ORT S5 15 DGHO

Page 1

Oncol Res Treat 38(suppl 5) XIV + 290 (2015)

38 | S5 | 15

print online ISSN 2296–5270 e-ISSN 2296–5262 ISBN 978-3-318-05626-6

www.karger.com/ort

E-Mail: jahrestagung2015@dgho-service.de

Jahrestagung der D­ eutschen, Österreichischen und ­Schweizerischen ­Gesellschaften für Hämatologie und ­Medizinische Onkologie Basel, 9.–13. Oktober 2015

ABSTRACTS Herausgeber

Martin Wernli, Aarau

S. Karger Medical and Scientific Publishers Basel · Freiburg · Paris · London · New York · Chennai · New Delhi · Bangkok · Beijing · Shanghai · Tokyo · Kuala Lumpur · Singapore · Sydney

Fotos: Basel Tourismus (3); Edition Phoenix Jutta Schneider, Michael Will

Band 38, Supplement 5, Oktober 2015


CONTENTS AUTHOR INDEX

Band 38, Supplement 5, Oktober 2015

Offizielles Organ von DGHO – Deutsche Gesellschaft für Hämatologie und Medizinische Onkologie OeGHO – Österreichische Gesellschaft für Hämatologie & Medizinische Onkologie DFaG – Deutsche Fatigue Gesellschaft AIO – Arbeitsgemeinschaft Internistische Onkologie in der Deutschen Krebsgesellschaft e.V. SGH-SSH – Schweizerische Gesellschaft für Hämatologie Mitglied der Deutschen Krebsgesellschaft e.V.

Editors Editor-in-Chief

Associate Editors

M. Hallek, Köln

S. Al-Batran, Frankfurt/M. C. Berking, München C. Bokemeyer, Hamburg M. Borner, Bern T. Cerny, St. Gallen H. T. Eich, Münster A. Engert, Köln M. Fassnacht, Würzburg F. Geiser, Bonn B. Groner, Frankfurt/M. V. Heinemann, München M. Hentrich, München R. D. Issels, München

W. Janni, Ulm U. R. Kleeberg, Hamburg A. A. Lammertsma, Amsterdam H. Lang, Mainz M. Moehler, Mainz P. Reichardt, Berlin M. Schuler, Essen R. Stupp, Zürich M. Theobald, Mainz R. Thomas, Köln U. Wedding, Jena J. A. Werner, Marburg O. Zivanovic, New York

A. Heidenreich, Aachen U. Herrlinger, Bonn A. Hochhaus, Jena R.-D. Hofheinz, Mannheim F. Honecker, St. Gallen R. D. Issels, München V. Jacobs, Salzburg K. Jordan, Halle U. Keilholz, Berlin J. P. Klussmann, Gießen H. Kölbl, Wien W. Kuhn, Bonn

H.-J. Lenz, Los Angeles P. Mallmann, Köln H. Moch, Zürich S. Reske, Ulm I. Runnebaum, Jena P. Schöffski, Leuven C. Spitzweg, München I. Strohscheer, St. Peter-Ording S. Ugurel, Essen R. Voltz, Köln M. Weller, Zürich

Editorial Board P. Albers, Düsseldorf C. Bausewein, München L. Bergmann, Frankfurt/M. J. Boos, Münster P. Brossart, Bonn W. Budach, Düsseldorf R. Büttner, Bonn J. Debus, Heidelberg E. Dippel, Ludwigshafen A. Du Bois, Essen T. Fehm, Düsseldorf N. Harbeck, München

Editorial Office S. Karger GmbH Attn. Dr. Steffi Hentzelt P.O. Box D-79095 Freiburg E-mail s.hentzelt@karger.com

Basel · Freiburg · Paris · London · New York · Chennai · New Delhi · Bangkok · Beijing · Shanghai · Tokyo · Kuala Lumpur · Singapore · Sydney


CONTENTS AUTHOR INDEX

Tagungspräsident

Dr. Martin Wernli, Aarau

Wissenschaftliches Sekretariat Prof. Dr. Mario Bargetzi, Aarau Prof. Dr. Christoph Mamot, Aarau Abstractgutachter Martin Adam, Bernd Alt-Epping, Reinhard Andreesen, Elisabeth Andritsch, Gabriela Baerlocher, Mario Bargetzi, Hannsjörg Baum, Walter Baumann, Dietrich W.  Beelen, Wolfgang Berdel, Lothar Bergmann, Jörg Beyer, Carsten Bokemeyer, Markus Borner, Martin Bornhäuser, Jan Braess, Tim Henrik Brümmendorf, Christian Buske, Marcelo Caballero, Nathan Cantoni, Jochen Casper, Yves Chalandon, Johannes Clausen, Antoinette Conca, Dieter Conen, Ivan Curjuric, Maike De Wit, Volker Diehl, Christian Dittrich, Peter Dreger, Ulrich Dührsen, Wilfried Eberhardt, Alexander Egle, Jutta Engel, Monika Engelhardt, Petra Fellmann, Paula Fernandez, Jürgen Finke, Thomas Fischer, Bernd Flath, Gunnar Folprecht, Mathias Freund, Michael Fridrik, Thomas Furrer, Arnold Ganser, Günther Gastl, Thomas Gauler, Klaus Geissler, Armin Gerger, Michele Ghielmini, Michael Girschikofsky, Hartmut Goldschmidt, Michael Gregor, Hildegard Greinix, Martin Grießhammer, Viktor Grünwald, Rainer Haas, Torsten Haferlach, Dominik Heim, Marc Heizmann, Daniel Helbling, Richard Herrmann, Viviane Hess, Pia Heußner, Wolfgang Hiddemann, Silvia Hofer, Florian Hohla, Ernst Holler, Friedemann Honecker, Andreas Huber, Gerald Illerhaus, Ulrich Jäger, Dirk Jäger, WolfDieter Janthur, Patrick Jermann, Karin Jordan, Ursula Kapp, Bernd Kasper, Felix Keil, Dieter Köberle, Gerald Kolb, Gabriela Kornek, Michael Krainer, Elena Kralidis, Nicolaus Kröger, Alois Lang, Florian Langer, Georg Lenz, Sabine Lenz, Sonja Loges, Andreas Lohri, Anja Lorch, Heinz Ludwig, Diana Lüftner, Andreas Mackensen, Christoph Mamot, Christine Mannhalter, Johannes Meran, Brigitte Mlineritsch, Peter Moosmann, Andreas Müller, Lothar Müller, Ralph Naumann, Dietger Niederwieser, Friedrich Overkamp, Ingrid Pabinger, Jens Panse, Jakob Passweg, Bernhard-C. Pestalozzi, Christina Peters, Andreas Petzer, Antonio Pezzutto, Michael Pfeilstöcker, Robert Pirker, Ferdinand Ploner, Matthias Preusser, Mirko Radloff, Peter Reichardt, Andreas Reiter, Christoph Renner, Oliver Rick, Hanno Riess, Ulrich Roelcke, Sacha Rothschild, Svetlana Sarinayova, Jan Schildmann, Richard F. Schlenk, Mathias Schmid, Norbert Schmitz, Alexander Schreiber, Ulrich Schuler, David Schwappach, Ulf Seifart, Marco Siano, Heinz Sill, Alexander Stein, Frank Stenner-Liewen, Roger Stupp, Georg Stüssi, Christian Taverna, Josef Thaler, Matthias Theobald, Christian Thiede, Michael Thomas, André Tichelli, Josef Trattner, Lorenz Trümper, Christian Urban, Philippe von Burg, Roger von Moos, Herbert Watzke, Ulrich Wedding, Rudolf Weide, Katja Weisel, ClemensMartin Wendtner, Martin Wernli, Martin Wilhelm, Wolfgang Willenbacher, Mathias WitzensHarig, Hans-Heinrich Wolf, Bernhard J. Wörmann, Walter A. Wuillemin, Stephen Wyler, Ralph Zachariah, Reinhard Zenhäusern, Nic Zerkiebel, Alfred Zippelius

Disclosure Statement The editor declares no conflict of interest.

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort


CONTENTS AUTHOR INDEX

Oncol Res Treat 2015;38(suppl 5)

Band 38, Supplement 5, Oktober 2015

Jahrestagung der D ­ eutschen, Österreichischen und ­Schweizerischen Gesellschaften für Hämatologie und Medizinische Onkologie Basel, 9.– 13. Oktober 2015

ABSTRACTS Herausgeber

Martin Wernli, Aarau

Die DGHO, OeGHO, SGMO und SGH übernehmen keine Gewähr für die Richtigkeit der Angaben in den Abstracts. Beiträge und Anzeigen geben nicht notwendigerweise die Auffassung der Vorstände wieder.

Basel · Freiburg · Paris · London · New York · Chennai · New Delhi · Bangkok · Beijing · Shanghai · Tokyo · Kuala Lumpur · Singapore · Sydney


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (chronologisch) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Expertenseminar

Primäre, chronische Immunthrombozytopenie

V6

1

Wissenschaftliches Symposium

ALL

V13

1

Fortbildung

Infektionen in der Onkologie und Hämatologie

V16–V18

2

Palliativmedizin interprofessionelle und interdisziplinäre Sichtweise

V22

2

Freier Vortrag

Pankreaskarzinom / Hepatobiliäres Karzinom / Melanom

V23–V28

4

Fortbildung

Ovarial- und Uteruskarzinom

V31

6

Freier Vortrag

AML experimentell I

V32–V37

7

Multiples Myelom klinisch

V38–V43

9

Lungenkarzinom I

V44–V49

13

Wissenschaftliches Symposium

Fortgeschrittenes Prostatakarzinom

V52

16

Fortbildung

Supportivtherapie bei neuen systemischen Therapieformen

V66

16

Nierenzellkarzinom – neue Aspekte und Standards

V67–V68

17

Freier Vortrag

Lymphome aggressiv klinisch I

V71–V75

17

Gesundheitspersonal neue Rollen, neue Aufgaben Fortbildung

V76

20

Freier Vortrag

MPN II / CML II

V80–V85

20

Wissenschaftliches Symposium

Long Term Survivorship

V86–V89

24

Freier Vortrag

Kopf-Hals-Tumore I

V90–V94

25

V96–V97

28

Hoch-Risiko Smouldering Myelom (SMM): Pro und Kontra unverzügliche Behandlung Wissenschaftliches Symposium Fortbildung

Sarkom – Vergleich der Behandlungskonzepte

V104–V105

29

Hodentumore – bestes Management

V106–V110

29

Freier Vortrag

MDS klinisch I

V114–V119

31

Palliativmedizin Entscheidungsprozesse und -kriterien Fortbildung

V121

33

Freier Vortrag

Allogene Transplantation klinisch III

V124–V129

34

Immuntherapie I

V130–V135

37

Expertenseminar

Genetische Beratung

V137

40

Wissenschaftliches Symposium

Kolon- und Rektumkarzinom Optimierungen der Strategien

V143

40

Fortbildung

HIV-assoziierte Tumoren

V147–V148

41

Wissenschaftliches Symposium

Maligne Gliome

V152–V153

41

Fortbildung

Kopf-, Hals-, Schild- und Speicheldrüsentumore – Update 2015

V154

42

Ethik Advance Care Planning

V159

42

Freier Vortrag

Allogene Transplantation klinisch I

V161–V166

43

V169

46

Leichtketten-Amyloidose: Aktueller Stand der Diagnostik und Therapie Expertenseminar Posterdiskussion

AML

P170–P187

46

Der spezielle Fall

P188–P199

53

Kolon-/Rektumkarzinom

P200–P214

57

Lymphome aggressiv

P215–P230

62

MDS / Stammzellen

P231–P244

68

Supportive Therapie

P245–P258

73

Urogenitale Malignome

P259–P273

80

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (chronologisch) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Fortbildung

MDS Differentialdiagnose und Therapiemodalitäten

V274–V276

87

Wissenschaftliches Symposium

NHL indolent neue Substanzen

V277–V279

87

Freier Vortrag

ALL

V285–V289

88

CLL klinisch

V290–V295

91

Fortbildung

Ethik Handeln am Lebensende

V296–V298

94

Freier Vortrag

Lymphome aggressiv experimentell

V299–V300

94

Tumor-/Zellbiologie I

V305–V310

98

Fortbildung

Hepatische Tumore

V313–V314

100

Expertenseminar

Stem Cell Biology

V316

101

Allogene Stammzelltransplantation

V317

101

Fortbildung

ALL

V319

101

Melanom neue Substanzen und Strategien

V324

102

Mammakarzinom Standards in der adjuvanten Behandlung

V326

102

Wissenschaftliches Symposium

Pankreaskarzinom

V331

102

Fortbildung

Urothelkarzinom – Management 2015

V334

102

Psychoonkologie

V339

103

Freier Vortrag

Stammzellen I

V341–V346

103

Nicht maligne Hämatologie

V350–V354

105

Expertenseminar

Porphyrie / Stoffwechselkrankheit

V359

107

Magenkarzinom

V360

108

Fortbildung

NHL aggressiv – Konzepte bei B- und T-Zell Lymphomen

V362

108

Wissenschaftliches Symposium

CLL I

V365–V367

109

Frühes Prostatakarzinom

V369–V371

109

Fortbildung

Geriatrische Onkologie

V377–V378

110

Wissenschaftliches Symposium

Stem Cell Biology – New Insights

V385–V386

111

Freier Vortrag

Kolon-/Rektumkarzinom I

V390–V395

111

Fortbildung

Transplantation Spendersuche GvHD Nachsorge

V399

114

AYA Adoleszente und junge Erwachsene – was ist anders?

V400–V402

114

Ösophaguskarzinom Therapiemodalitäten State of the Art

V403–V406

115

Patientensicherheit

V408–V410

116

Hämostaseologie II

V412–V413

117

Freier Vortrag

Lymphome indolent

V418–V423

118

AML klinisch

V424–V429

121

Mammakarzinom I

V430–V435

124

Posterdiskussion

Allogene Transplantation

P436–P446

126

CML

P447–P458 120

Lungenkarzinom

P459–P468

136

Lymphatische Neoplasien und nicht-maligne Hämatologie

P469–P482

140

Querschnittsthemen I

P483–P493

145

Sonstige Themen I

P494–P503

150

Versorgungsforschung

P504–P519

153

Wissenschaftliches Symposium

Lunge / Pleura personalisierte Therapieansätze

V521

159

Fortbildung

MPN Chronische Myeloproliferative Erkrankungen

V526–V527

160

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (chronologisch) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Konfrontation oder Kooperation: Interaktion von Onkologen und Organonkologen

V533

160

Freier Vortrag

AML experimentell II

V537–V542

160

Stammzellen II

V543–V546

163

Multiples Myelom experimentell

V547–V552

164

GIST / Urogenitale Malignome / sonstige Tumore

V553–V558

167

Immuntherapie II

V559–V564

169

Patientenberatung und Krebsregister

V565–V569

172

Wissenschaftliches Symposium

Immunphänotypisierung

V571

174

Plenarsitzung

Best Abstract

V579–V584

174

Wissenschaftliches Symposium

Multiples Myelom Risiko- und altersadaptiertes Vorgehen

V585–V588

177

Fortbildung

NHL indolent unterschiedliche Krankheiten – Gemeinsamkeiten

V589–V591

178

Wissenschaftliches Symposium

Metastasiertes Mammakarzinom

V597

179

Freier Vortrag

CML I

V600–V605

179

MDS klinisch II

V606–V611

183

Kopf-Hals-Tumore II

V612–V615

186

Wissenschaftliches Symposium

Young Investigator‘s Award Preisverleihung

V616–V621

187

Expertenseminar

Früherkennung Lungenkarzinom

V622

189

Freier Vortrag

Supportive Therapie I

V625–V630

190

Tumor-/Zellbiologie II

V631–V636

193

Wissenschaftliches Symposium

AML – Neue Substanzen

V637–V638

195

Fortbildung

Intensivmedizin

V641

195

Wissenschaftliches Symposium

MDS Zellregulation

V644

196

Supportive Therapien

V649–V650

196

Freier Vortrag

Lymphome aggressiv klinisch II

V655–V660

197

CLL experimentell

V661–V666

199

Mammakarzinom II / Nierenzellkarzinom

V670–V675

202

Expertenseminar

Ovarialkarzinom fortgeschritten

V680

205

Pankreaskarzinom

V681

205

Freier Vortrag

Kolon-/Rektumkarzinom II

V682–V686

205

Translationale Forschung

V687–V692

207

Fortbildung

Magenkarzinom

V694–V696

210

Wissenschaftliches Symposium

Transplantation

V698

210

Freier Vortrag

Hodgkin-Lymphome / B- und T-Zell-Lymphome

V704–V708

211

Wissenschaftliches Symposium

DGTI Joint Symposium: Neuartige Zelltherapien

V709–V711

212

Freier Vortrag

AML experimentell III

V714–V719

213

MPN I

V720–V725

215

Palliativmedizin

V730–V735

218

Supportive Therapie II

V742–V747

221

Versorgungsforschung

V748–V753

223

Posterdiskussion

MPN / sonstige Hämatologie

P754–P768

226

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (chronologisch) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Multiples Myelom I

P769–P780

233

Multiples Myelom II

P781–P791

237

Querschnittsthemen II

P792–P802

241

Sarkom

P803–P811

245

Sonstige Themen II

P812–P820

248

Verschiedene solide Tumore

P821–P831

251

Wissenschaftliches Symposium

CLL II

V832–V835

256

Freier Vortrag

Immuntherapie III

V846–V851

257

MDS experimentell

V852–V857

259

Allogene Transplantation klinisch II

V858–V863

262

Wissenschaftliches Symposium

PET bei Lymphomen

V868–V869

264

V872

265

Kompetenznetzwerk Leukämien – Neue Erkenntnisse bei AML/ALL/CML/MDS Fortbildung

Freier Vortrag

Prostatakarzinom

V878–V883

265

Lungenkarzinom II

V884–V889

268

Author Index Imprint

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

272 290


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (nach Sitzung) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Plenarsitzung

Best Abstract

V579–V584

Wissenschaftliches Symposium

ALL

V13

AML – Neue Substanzen

V637–V638

195

CLL I

V365–V367

109

CLL II

V832–V835

256

DGTI Joint Symposium: Neuartige Zelltherapien

V709–V711

212

Fortgeschrittenes Prostatakarzinom

V52

Frühes Prostatakarzinom

V369–V371

Hoch-Risiko Smouldering Myelom (SMM): Pro und Kontra unverzügliche Behandlung

V96–V97

174 1

16 109 28

Immunphänotypisierung

V571

174

Kolon- und Rektumkarzinom Optimierungen der Strategien

V143

40

Long Term Survivorship

V86–V89

24

Lunge / Pleura personalisierte Therapieansätze

V521

Maligne Gliome

V152–V153

MDS Zellregulation

V644

196

Metastasiertes Mammakarzinom

V597

179

Multiples Myelom Risiko- und altersadaptiertes Vorgehen

V585–V588

177

NHL indolent neue Substanzen

V277–V279

87

Pankreaskarzinom

V331

102

PET bei Lymphomen

V868–V869

264

Stem Cell Biology – New Insights

V385–V386

111

Supportive Therapien

V649–V650

196

Transplantation

V698

210

Young Investigator‘s Award Preisverleihung

V616–V621

187

Fortbildung

ALL

V319

101

AYA Adoleszente und junge Erwachsene – was ist anders?

V400–V402

114

Ethik Advance Care Planning

V159

42

Ethik Handeln am Lebensende

V296–V298

94

Geriatrische Onkologie

V377–V378

110

Gesundheitspersonal neue Rollen, neue Aufgaben

V76

159 41

20

Hämostaseologie II

V412–V413

117

Hepatische Tumore

V313–V314

100

HIV-assoziierte Tumoren

V147–V148

41

Hodentumore – bestes Management

V106–V110

29

Infektionen in der Onkologie und Hämatologie

V16–V18

Intensivmedizin

V641

195

Kompetenznetzwerk Leukämien – Neue Erkenntnisse bei AML/ALL/CML/MDS

V872

265

Konfrontation oder Kooperation: Interaktion von Onkologen und Organonkologen

V533

160

Kopf-, Hals-, Schild- und Speicheldrüsentumore – Update 2015

V154

Magenkarzinom

V694–V696

2

42 210


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (nach Sitzung) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Mammakarzinom Standards in der adjuvanten Behandlung

V326

MDS Differentialdiagnose und Therapiemodalitäten

V274–V276

Melanom neue Substanzen und Strategien

V324

102

MPN Chronische Myeloproliferative Erkrankungen

V526–V527

160

NHL aggressiv – Konzepte bei B- und T-Zell Lymphomen

V362

108

NHL indolent unterschiedliche Krankheiten – Gemeinsamkeiten

V589–V591

178

Nierenzellkarzinom – neue Aspekte und Standards

V67–V68

Ösophaguskarzinom Therapiemodalitäten State of the Art

V403–V406

Ovarial- und Uteruskarzinom

V31

6

Palliativmedizin Entscheidungsprozesse und -kriterien

V121

33

Palliativmedizin interprofessionelle und interdisziplinäre Sichtweise

V22

2

102 87

17 115

Patientensicherheit

V408–V410

116

Psychoonkologie

V339

103

Sarkom – Vergleich der Behandlungskonzepte

V104–V105

Supportivtherapie bei neuen systemischen Therapieformen

V66

16

Transplantation Spendersuche GvHD Nachsorge

V399

114

Urothelkarzinom – Management 2015

V334

102

Expertenseminar

Allogene Stammzelltransplantation

V317

101

Früherkennung Lungenkarzinom

V622

189

Genetische Beratung

V137

40

V169

46

Leichtketten-Amyloidose: Aktueller Stand der Diagnostik und Therapie

29

Magenkarzinom

V360

108

Ovarialkarzinom fortgeschritten

V680

205

Pankreaskarzinom

V681

205

Porphyrie / Stoffwechselkrankheit

V359

107

Primäre, chronische Immunthrombozytopenie

V6

Stem Cell Biology

V316

Freier Vortrag

Pankreaskarzinom / Hepatobiliäres Karzinom / Melanom

V23–V28

ALL

V285–V289

Allogene Transplantation klinisch I

V161–V166

43

Allogene Transplantation klinisch II

V858–V863

262

Allogene Transplantation klinisch III

V124–V129

34

AML experimentell I

V32–V37

AML experimentell II

V537–V542

160

AML experimentell III

V714–V719

213

AML klinisch

V424–V429

121

CLL experimentell

V661–V666

199

CLL klinisch

V290–V295

91

CML I

V600–V605

179

GIST / Urogenitale Malignome / sonstige Tumore

V553–V558

167

Hodgkin-Lymphome / B- und T-Zell-Lymphome

V704–V708

211

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

1 101 4 88

7


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (nach Sitzung) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

Immuntherapie I

V130–V135

37

Immuntherapie II

V559–V564

169

Immuntherapie III

V846–V851

257

Kolon-/Rektumkarzinom I

V390–V395

111

Kolon-/Rektumkarzinom II

V682–V686

205

Kopf-Hals-Tumore I

V90–V94

Kopf-Hals-Tumore II

V612–V615

Lungenkarzinom I

V44–V49

Lungenkarzinom II

V884–V889

268

Lymphome aggressiv experimentell

V299–V300

94

Lymphome aggressiv klinisch I

V71–V75

Lymphome aggressiv klinisch II

V655–V660

197

Lymphome indolent

V418–V423

118

Mammakarzinom I

V430–V435

124

Mammakarzinom II / Nierenzellkarzinom

V670–V675

202

MDS experimentell

V852–V857

259

MDS klinisch I

V114–V119

31

MDS klinisch II

V606–V611

183

MPN I

V720–V725

215

MPN II / CML II

V80–V85

Multiples Myelom experimentell

V547–V552

Multiples Myelom klinisch

V38–V43

Nicht maligne Hämatologie

V350–V354

105

Palliativmedizin

V730–V735

218

Patientenberatung und Krebsregister

V565–V569

172

Prostatakarzinom

V878–V883

265

Stammzellen I

V341–V346

103

Stammzellen II

V543–V546

163

Supportive Therapie I

V625–V630

190

Supportive Therapie II

V742–V747

221

Translationale Forschung

V687–V692

207

Tumor-/Zellbiologie I

V305–V310

98

Tumor-/Zellbiologie II

V631–V636

193

Versorgungsforschung

V748–V753

223

Posterdiskussion

AML

P170–P187

46

Allogene Transplantation

P436–P446

126

CML

P447–P458 120

Der spezielle Fall

P188–P199

53

Kolon-/Rektumkarzinom

P200–P214

57

Lungenkarzinom

P459–P468

136

Lymphatische Neoplasien und nicht-maligne Hämatologie

P469–P482

140

Lymphome aggressiv

P215–P230

62

MDS / Stammzellen

P231–P244

68

25 186 13

17

20 164 9


CONTENTS AUTHOR INDEX

Contents ∙ Inhalt ∙ (nach Sitzung) Oncol Res Treat 2015;38(suppl 5)

Abstract-No. Page-No.

MPN / sonstige Hämatologie

P754–P768

226

Multiples Myelom I

P769–P780

233

Multiples Myelom II

P781–P791

237

Querschnittsthemen I

P483–P493

145

Querschnittsthemen II

P792–P802

241

Sarkom

P803–P811

245

Sonstige Themen I

P494–P503

150

Sonstige Themen II

P812–P820

248

Supportive Therapie

P245–P258

73

Urogenitale Malignome

P259–P273

80

Verschiedene solide Tumore

P821–P831

251

Versorgungsforschung

P504–P519

153

Author Index Imprint

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

272 290


CONTENTS AUTHOR INDEX

Abstracts Oncol Res Treat 2015;38(suppl 5):1–270 DOI: 10.1159/000439070

Expertenseminar

Primäre, chronische Immunthrombozytopenie

Wissenschaftliches Symposium ALL

V6

V13

Primary, chronic immunethrombocytopenia

Clonal evolution in acute lymphoblastic leukemia – new insights via high-throughput sequencing and MRD analyses

Imbach P.1 Universität Basel, Uettligen, Switzerland

1

Introduction: Within the last 3 decades the knowledge of the history, demographics, immune pathology, clinical presentation and management dramatically have been expanded. Methods: Prospecive registries, guidelines, the characteristics of the disturbed immune response by laboratory and clinical studies and new modalities of management are the main factors of improved knowledges. Results: The prospective registries of immune thrombocytopenia led to new terminologies. Since only a part of patients with chronic ITP has symptoms of bleeding thrombocytopenic “Purpura” is no longer in use. The pathogenetic alterations of the innate and the adaptive immune system and the immune modulatory effects of new treatment modalities are the reasons to adapt the “i” from idiopathic to immune or autoimmune thrombocytopenia.In addition studies of the amount of platlet counts between 100–150 K showed that less than 7 percents of individuals developed ITP during further observation times. Follow up data of prospective registries with high numbers of patients revealed spontaneous recovery of up to 25% of patients between 6–12 months after diagnosis. Thus the terme “chronic ITP “ has been postponed to 12 months after diagnosis. It was recognized that patients with no or mild bleeding management often do not need active treatment, but these patients have to be strictly followed for improvement of their quality of life. Patients with acute and/or severe bleeding need intravenous immunoglobulins IVIG, high dose steroids and rarely platelet transfusions. The guidelines of definitions and managements resulted in more individualized management including the quality of life of the patient. Special considerations of management are needed in hyperactive children, in elderly persons with higher rates of comorbities, in pregnant womem, especially during labor an delivery, and in patients, who need preventive treatment (i.e befory surgery, acive sports etc.). The management may be observation only in patient with no or mild bleeding. Active treatment modalities are now turning from the classic forms with high risk of adverse effects to acceptable side efffects (i.e. rituximab) and treatments with low adverse effects and higher rates of responses (i.e.in patients with bleeding. IVIG, in patients at risk of bleeding: thrombopoetin receptor agonists). Conclusions: Registries may help to define special risk groups of patients with chronic ITP, which then should be evaluated by controlled studies. Disclosure: No conflict of interest disclosed.

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

Brüggemann M.1 Universitätsklinikum Schleswig-Holstein, Campus Kiel, Medizinische Klinik II, Kiel, Germany 1

Acute lymphoblastic leukemia (ALL) is regarded as monoclonal disease deriving from an uncontrolled proliferation of a transformed lymphoid precursor cell. However, already early studies on immunoglobuline (IG) and T-cell receptor (TR) gene rearrangement patterns in ALL for MRD analyses challenged the concept of monoclonality as recurrently the number of detectable clonal rearrangements exceeds the maximum number of rearrangements within one clone and differences in rearrangements patterns are detected between initial diagnosis and relapse. Conventional treatment concepts are generally not affected by subclone detection because the diagnostic workup focuses on cytogenetic, molecular and immunophenotypic characteristics of the leukemic bulk with these factors being partially used for risk stratification of ALL. However, our increased understanding of molecular mechanisms of cancer and availability of drugs targeting them is changing the meaning of subclonal aberrations and their evolution in the diagnostics, treatment and follow-up of ALL. Next generation sequencing (NGS) of amplified IG/TR gene rearrangements allows sensitive MRD assessement of leukemia associated IG/TR gene rearrangements and gives insights into composition of additional low frequency subclones. Published studies identified up to thousands of different subclones per patients using this method. Also within the GMALL a series of diagnostic and relapse ALL samples were investigated using IG/TR NGS demonstrating high levels of oligoclonality. However, within this limited series none of the minor subclones at diagnosis became dominant at relapse so that the meaning of these subclones remains to be elucidated. Molecular analyses of other recurrent genetic alterations like intrageneic deletions of the BTG1, IKZF1 and ERG confirm ALL clonal variegation also for these aberrations. If molecular aberrations serve as treatment targets monitoring of these targets can identify clonal selection processes. If e. g. NUP214-ABL1 positive T-ALL is treated with tyrosine kinase inhibitors selection of NUB214-ABL1 negative subclones is a possible mechanism of resistance. Also low-level resistance mutations like BCR-ABL tyrosine kinase domain mutations in newly diagnosed or recurrent BCR-ABL positive ALL may play a future role for choice of tyrosine kinase inhibitors. Therefore, sensitive mutation detection strategies to monitor driver mutations or druggable targets may complement the analysis of bulk MRD. Disclosure: No conflict of interest disclosed.


CONTENTS AUTHOR INDEX

Fortbildung

Infektionen in der Onkologie und Hämatologie V16

Diagnosis of fungal infections – current guideline 2015 Schwartz S.1 Charité Campus Benjamin Franklin, Medizinische Klinik mit Schwerpunkt Hämatologie und Onkologie, Berlin, Germany 1

Invasive fungal infections (IFI) remain a diagnostic challenge, although progress with development of new tools has been made. To establish a definite diagnosis of an IFI, growth of the infecting fungus from an otherwise sterile site or histopathological or microscopic demonstration of the fungus with evidence of tissue invasion or damage or microscopy with characteristic morphological findings is usually required. This is of particular importance in growing numbers of patients with infections caused by rare, emerging fungi. However, invasive procedures are not always feasible due to frequent coagulation impairment in immunocompromised patients. Imaging (eg, high-resolution CT-scan) and serological techniques (eg, galactomannan) are well established, but, with few exceptions (eg, cryptococcosis), findings support a probable diagnosis and are not sufficient to establish a definite diagnosis. Fungus-specific PCR-techniques, either targeting pan-fungal or genus-specific gene sequences, could detect presence of fungal nucleic acids in blood specimens or bronchoalveolar lavage (BAL). This technique displays an enhanced sensitivity and specificity, particularly when BAL samples are analysed. Combination of results from PCR and serological studies could further enhance the diagnostic sensitivity and specificity. However, various PCR techniques exist and there are no commonly accepted protocols, which currently precludes incorporation of this technique as mandatory into diagnostic algorithms. A new, innovative diagnostic tool, which detects volatile, fungus-specific compounds in breath samples from patients with pulmonary aspergillosis, has been recently developed. This technique not only has the potential to be used as a rapid bedside test, but to distinguish infections with different fungi. Another novel approach uses detection of fungus-specific, CD4-positive T-cells in blood samples. The sensitivity of this test is enhanced by brief in vitro stimulation with fungal antigens and subsequent detection of CD154 up-regulation by flow cytometry. This diagnostic approach is somewhat limited by large volumes of diagnostic blood specimens, but could differentiate between infections with various types of fungi. In conclusion, the updated guideline about the diagnosis of fungal infections will not only gather new data about established diagnostic tools, but incorporate new, innovative diagnostic tools. Disclosure: Stefan Schwartz: Advisory Role: Pfizer Pharma GmbH, BTG International Ltd, AMGEN; Financing of Scientific Research: MSD Sharp & Dohme, Pfizer Pharma GmbH, Gilead Sciences GmbH, Astellas GmbH, AMGEN. V18

Emergence of multi-resistant bacteria in hematological and oncological settings Biehl L.M.1,2 Klinik I für Innere Medizin, Uniklinik, Köln, Germany, 2Deutsches Zentrum für Infektionsforschung (DZIF), Bonn-Köln, Germany 1

The prevalence of multi-resistant bacteria (MRB), in particular gram negative bacteria, is increasing worldwide. While these bacteria are most frequently found colonizing the intestinal tract, they can cause invasive infections especially in immunocompromised patients. Due to the limited therapeutic options and the frequent delay in adequate treatment, infections are associated with an increased mortality as compared to non multi-resistant pathogens. The epidemiology of extended-spectrum ß-lactamase producing Enterobacteriaceae (ESBL-E) and carbapenem-resistant gram negative bacteria is highly depending on regional factors and charateristics of the popula-

2

Oncol Res Treat 2015;38(suppl 5):1–270

tion under scrutiny. So far, carbapenem-resistance is still infrequent in Germany and Northern Europe. Certain risk factors including previous antibiotic treatment and medical care in high-risk countries have been identified. In settings with hematological and oncological patients, a screening strategy based on the local epidemiology should be considered. Furthermore, individualized adaption of empirical antibiotic treatment in febrile neutropenia depending on colonization status may be beneficial. However, this needs to be implemented with caution to avoid overuse of broad-spectrum antibiotcs and further selection pressure. Disclosure: No conflict of interest disclosed.

Fortbildung

Palliativmedizin interprofessionelle und interdisziplinäre Sichtweise V22

The view on palliative medicine from different perspectives: Palliative care, medical oncology, internal medicine Lordick F.1 Universitäres Krebszentrum Leipzig (UCCL), Leipzig, Germany

1

6 years ago ASCO released a statement that was adopted by the Arbeitsgemeinschaft Palliativmedizin of the German Cancer Society: “Palliative cancer care is the integration into cancer care of therapies that address the multiple issues that cause suffering for patients and their families and impact their life quality. Effective provision of palliative cancer care requires an interdisciplinary team that can provide care in all patient settings, including outpatient clinics, acute and long-term care facilities, and private homes. Changes in current policy, drug availability, and education are necessary for the integration of palliative care throughout the experience of cancer, for the achievement of quality improvement initiatives, and for effective palliative cancer care research. The need for palliative cancer care is greater than ever notwithstanding the strides made over the last decade. Further efforts are needed to realize the integration of palliative care in the model and vision of comprehensive cancer care by 2020.” The publication of the German multidisciplinary and multiprofessional evidence-based and expert-consented (S3) “Guideline for Palliative Care of Patients with Incurable Cancer” is an important step into this direction. Another milestone shall be the planned law for improving hospice and palliative care in Germany (Hospiz- und Palliativgesetz HPG) which is in preparation. In the past, research data in the field of palliative care were scarce. Actions were taken on a more intuitive and empirical basis. Meanwhile, important research results have been published elucidating for instance the role of early integration of palliative care into routine oncology. This raises the question who should deliver palliative care to cancer patients. First and foremost, we need to better understand the real burden on our patients, their preferences and care needs during the course of an incurable disease. This is why we started a multidisciplinary research program in the scientific working group (Sektion B) of the German Cancer Society. Eventually, we need to tailor “who shall provide” and “how to deliver” optimized palliative care. The current understanding is that every physician who cares for patients with severe and incurable diseases should have been trained and should be able to deliver general aspects of symptom relieve and palliative care. For more complicated and extended palliative and “end-of-life” care, specialized teams are required. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag

V24

Pankreaskarzinom / Hepatobiliäres Karzinom / Melanom V23

The pancreatic carcinoma – treatment research and treatment reality in oncology practices Weber K.1, Schlichting A.2, Tessen H.-W.3,4 Onkologische Praxis Hellersdorf, Berlin, Germany, 2rgb Onkologisches Management GmbH, Sarstedt, Germany, 3Onkologische Kooperation Harz, Goslar, Germany, 4Projektgruppe Internistische Onkologie (PIO), Goslar, Germany 1

Introduction: According to the 2013-S3 guideline “Exocrine pancreatic carcinoma“, surgery is the only curative treatment option for pancreatic carcinoma. Gemcitabine, gemcitabine/ erlotinib, FOLFIRINOX are recommended as adjuvant or palliative 1st-line therapy, 5-FU/oxaliplatin as 2nd-line therapy. Methods: Data related to the treatment of pancreatic cancer have been analysed within the national scientific register ONCOReg since March 2009. The register contains the records of a total of 30,186 patients from 355 practices in 15 federal states, including 1,722 pancreatic carcinomas from 66 practices (2,542 therapies). 1,522 disease histories are available for evaluation so far. Results: Patient characteristics: Gender: 821 (53.9%) male, 701 (46.1%) female Age at initial diagnosis: 69 (35–95) year; 22% older than 75 years UICC stages: 43 (2.8%) I; 540 (35.5%) II; 154 (10.1%) III; 698 (45.9%) IV, 88 (5.8%) n.k. 634 (41.7%) patients were operated. An R0 resection was achieved in 491 cases (77.4%), an R1 resection in 109 (17.2%) patients. Adjuvant therapies: 420 (66.2%) patients, R1-additive therapies: 90 (14.2%); 490 (96.1%) gemcitabine monotherapy. Duration of treatment at 5.1 resp. 4.7 months. Survival rates of the adjuvant resp. R1-additive therapy: DFS 14.1 resp. 9.1 months; OS 33.6 resp. 18.8 months. Palliative therapies: 1,268 patients received 1,978 palliative therapies: 726 (57.3%) gemcitabine; 371 (29.3%) gemcitabine/erlotinib; 138 (10.9%) FOLFIRINOX; 130 (10.3%) OFF; 88 (7.0%) FUFOX, 68 (5.4%) gemcitabine/albumin-bound paclitaxel; 54 (4.3%) gemcitabine/oxaliplatin. The median duration of treatment was at 78 days (91 days for 1st-line therapies). 1,268 patients received a 1st-line therapy; 519 (41.0%) a 2nd-line; 143 (11.3%) a 3rd-line therapy. The responses were assessed in 1,846 therapies, and the objective response (CR/PR) was at 9.9%. In 34.0% of the cases a halt of the progression of the disease was achieved. A progression had to be observed in 36.3%, with the PD rate increasing with increasing therapy line (1st-line 32.6%; 2nd-line 43.3%; 3rd-line 50.0%). Survival data: PFS: 1st-line 4.9 mths.; 2nd-line 3.3 mths. OS: 1st-line 9.0 mths.; 2nd-line 6.2 mths. Conclusions: Data collection and analysis is an integral part of the routine in oncology practices.Data on the duration of therapy, the response and survival of selected first- and second-line therapies will be presented. Disclosure: No conflict of interest disclosed.

4

Oncol Res Treat 2015;38(suppl 5):1–270

Transduction with C-C-chemokine receptor type 4 (CCR4) enhances tumor-specific migration of adoptively transferred T cells in a model of pancreatic cancer Rapp M.1, Grassmann S.1, Endres S.1, Anz D.1, Kobold S.1 Ludwig-Maximilians-Universität, Abteilung für Klinische Pharmakologie, München, Germany 1

Introduction: Regulatory T cells selectively express C-C-chemokine receptor type 4 (CCR4) and are attracted by intratumoral CCL22 while CCR4 is absent from most cytotoxic T cells. We hypothesized that inducing forced expression of CCR4 on adoptively transferred cytotoxic T cells could enhance therapeutic efficacy. Methods: CCR4 and a new non-functional CCR4-deletion mutant were cloned into the retroviral vector pMP71 and transduced into murine ovalbumin-specific T cells (OT-1). T cell migration and function in vitro were assessed using transwell assays. In vivo efficacy was assessed in a murine model induced with the syngeneic pancreatic cell line Panc02-OVA. Results: In vitro, CCR4- but not CCR4del-transduced OT-1 T cells specifically migrated towards recombinant CCL22 and tumor-induced CCL22 (25-fold increase). In vivo, CCR4-transduced OT-1 T cells cured 50% of mice bearing established Panc-OVA tumors compared to 12% in the CCR4del- or untransduced OT-1 T cell- treated mice (n = 8 per group, p < 0.0001). Cured mice were protected from rechallenge, indicating the induction of anti-tumor immunity. Enhanced numbers of CCR4-transduced OT-1 T cells compared to CCR4del-transduced cells were found in the tumor, suggesting enhanced homing to the tumor site as one mode of action of CCR4-transduced T cells. Conclusions: The transduction of cytotoxic T cells with CCR4 represents a new therapeutic approach for effectively guiding adoptively transferred T cells into pancreatic tumors and thereby inducing a potent antitumor immune response. Disclosure: No conflict of interest disclosed. V25

Evaluation of systemic inflammatory response (SIR) markers in pancreatic ductal adenocarcinoma (PDAC) Markus M.1, Kasper S.1, Noureddine R.1, Abramczyk M.1, Paul A.2, Gerken G.3, Schmid K.W.4, Markus P.5, Schumacher B.6, Meiler J.1, Wiesweg M.1, Kaiser G.2, Dechêne A.3, Trarbach T.7, Schuler M.1, Abendroth A.1 Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Innere Klinik (Tumorforschung), Essen, Germany, 2Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Essen, Germany, 3Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Gastroenterologie und Hepatologie, Essen, Germany, 4Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Institut für Pathologie, Essen, Germany, 5Elisabeth Krankenhaus Essen, Klinik für Allgemein-, Viszeral- und Unfallchirurgie, Essen, Germany, 6Elisabeth Krankenhaus Essen, Klinik für Gastroenterologie, Essen, Germany, 7iOMEDICO AG, Freiburg, Germany 1

Introduction: Intratumoral inflammation is a known hallmark of cancer and can promote tumorigenesis especially in PDAC. Recently, anti-inflammatory agents like JAK2- or CCR2-inhibitors showed promising results in early clinical trials in patients with metastatic PDAC. So far, valid biomarkers for the response prediction to these novel anti-inflammatory drugs are missing but essential, as only a subset of patients seem to respond. We retrospectively evaluated the prognostic and predictive value of routinely assessed inflammatory serum response (SIR) biomarkers in patients with advanced PDAC treated with systemic chemotherapy at the West German Cancer Center (WTZ) in Essen, one of the 13 Oncology Centers of Excellence in Germany. Methods: A total of 245 PDAC patients treated at the WTZ from 2005– 2012 were retrospectively evaluated for the SIR markers: Glasgow Prognostic Score (GPS, mGPS), C-reactive-Protein (CRP), Neutrophile-/ Lymphocyte Ratio (NLR), Lymphocyte-Monocyte Ratio (LMR) and

Abstracts


CONTENTS AUTHOR INDEX

Platelet-/Lymphocyte Ratio (PLR). SIR markers were correlated with clinico-pathological findings, response to chemotherapy and overall survival (OS) using Kaplan-Meier curves, univariate and multivariate Cox proportional models. Results: In metastatic PDAC median OS was 8.5 months (95% CI 6.95– 10.35). All evaluated SIR markers except of PLR were significantly associated with reduced OS (Figure 1). The HR and (95% CI) for PLR>200 were: 0.829 (0.512–1.309, p = 0.403), for NLR< 5: 1.899 (1.175–3.069, p = 0.009), for LMR< 2.8: 1.926 (1.111–3.338, p = 0.019), for CRP>normal: 2.536 (1.568–4.101, p < 0.001), for GPS>2: 3.266 (1.269–8.407, p = 0.014) and for mGPS: 1.993 (1.010–3.930, p = 0.047). In patients receiving gemcitabine monotherapy (48.8%) the evaluated SIR markers had no impact on progression free survival (PFS), but in patients receiving combination chemotherapy (51,2%, mostly gemcitabine/cisplatin or oxaliplatin) most evaluated SIR markers correlated with reduced PFS. Conclusion: SIR biomarkers, routinely assessed in clinical settings can be easily used as prognostic and predictive markers to optimize therapeutic strategies in PDAC.

pts and tumor characteristics, comorbidities, factors affecting therapeutic decision making, previous and ongoing systemic treatments as well as outcome data such as response, progression free (PFS) and overall survival data are recorded. Here, characteristics and first-line treatment of pts with advanced or metastatic pancreatic cancer are reported for the first 187 pts recruited into the registry. In addition, outcome data such as firstline PFS and six-month survival rate are presented. Results: At the start of palliative treatment pts mean age was 69 years (yrs), 55% of pts were male. 73% of pts had metastatic disease. Palliative 1st-line treatment was generally based on gemcitabine in combination with nab-paclitaxel (GEM+Nab-Pac, 42%), gemcitabine monotherapy (GEM mono, 27%) and less frequently on FOLFIRINOX (18%). Pts receiving GEM mono are older and have more comorbidities than pts receiving GEM+Nab-Pac or FOLFIRINOX (mean age 74 yrs vs. 69 and 61 yrs; mean Charlson comorbidity score 1.1 vs. 0.4 and 0.3). After a median follow-up of 7.6 months, median PFS of all pts is 5.3 months (95% CI: 4.7–6.2). Overall survival rate at 6 months is 62%. Conclusion: In routine practice, pts with metastatic or advanced pancreatic cancer are treated according to age and comorbidities. Although pts in routine practice are markedly older than pts in clinical trials, median PFS is promising. Further analysis will investigate prognosis factors and the effectiveness of different treatment options. Disclosure: No conflict of interest disclosed. V27

Evaluation of systemic inflammatory response (SIR) markers in advanced biliary tract cancer (ABTC) Abendroth A.1, Markus M.1, Abramczyk M.1, Noureddine R.1, Paul A.2, Gerken G.3, Schmid K.W.4, Markus P.5, Schumacher B.6, Meiler J.1, Wiesweg M.1, Kaiser G.2, Dechêne A.3, Trarbach T.7, Schuler M.1, Kasper S.1 Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Innere Klinik (Tumorforschung), Essen, Germany, 2Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Allgemein,- Viszeral- und Transplantationschirurgie, Essen, Germany, 3Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Gastroenterologie und Hepatologie, Essen, Germany, 4Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Institut für Pathologie, Essen, Germany, 5Elisabeth Krankenhaus Essen, Klinik für Allgemein,- Viszeral- und Unfallchirurgie, Essen, Germany, 6Elisabeth Krankenhaus Essen, Klinik für Gastroenterologie, Essen, Germany, 7iOMEDICO AG, Freiburg, Germany 1

Fig. 1. Hazard Ratio for Overall survival Disclosure: No conflict of interest disclosed.

Introduction: Pancreatic cancer is the fourth leading cause of cancer death in Europe. About 80% of patients (pts) are diagnosed with locally advanced or metastatic disease and with a 5-year survival rate of less than 2%. Thus, pancreatic cancer remains a disease with dismal prognosis. Within the last decade new treatment options have become available. Clinical registries provide insight into treatment reality and real-life effectiveness outside of clinical trials. Methods: The Tumor Registry Pancreatic Cancer (TPK) is a prospective, multicenter, observational study of pts with advanced or metastatic pancreatic cancer receiving systemic antineoplastic palliative therapy. Since February 2014, 99 study sites in Germany have recruited more than 400 pts at the start of their first-line treatment. A broad set of data regarding

Introduction: Systemic inflammatory response (SIR) markers are prognostic in several cancers but have not been evaluated systematically in large ABTC series. We evaluated the prognostic and predictive value of the modified Glasgow Prognostic Score (mGPS), C-reactive protein (CRP), Neutrophile-/Lymphocyte Ratio (NLR), Lymphocyte-Monocyte Ratio (LMR) and Platelet-/Lymphocyte Ratio (PLR) in patients with ABTC treated with systemic chemotherapy at the West German Cancer Center (WTZ) in Essen, one of the 13 Oncology Centers of Excellence in Germany. Methods: SIR markers were retrospectively evaluated in 147 patients (pts) with ABTC treated with palliative systemic chemotherapy at the WTZ from 2005–2012. Cutoff values for PLR, NLR and LMR defining high vs. low scores were selected by rounding the mean values for the entire cohort, which were 218, 4.6 and 2.6, respectively. For CRP, values above 10mg/dl were selected. The mGPS is based on CRP and albumin. Pts who had both elevated CRP (>10mg/l) and hypoalbuminaemia (< 35g/l) had a score of 2. SIR markers were correlated with clinico-pathological findings, response to chemotherapy, progression free (PFS) and overall survival (OS) using Kaplan-Meier curves and Cox proportional hazard models. Results: In ABTC median OS was 13.9 months (95% CI 10.5–17.4). All evaluated SIR markers were significantly associated with reduced OS (table 1). Interestingly, in pts receiving combination chemotherapies (78.8%, mostly gemcitabine/oxaliplatin or cisplatin) none of the evaluated SIR markers had an impact on PFS or OS. In contrast pts receiving monochemotherapy (21.2%, mostly gemcitabine) SIR markers strongly correlated with reduced PFS and OS.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

V26

Progression-free and overall survival of patients with advanced or metastatic pancreatic cancer in German outpatient centers – first outcome data from the clinical TPK Registry Hegewisch-Becker S.1, Hofheinz R.2, Wolf T.3, Aldaoud A.4, Harde J.5, Kopfmann S.5, Marschner N.6 Onkologische Schwerpunktpraxis, Hamburg, Germany, 2Universitätsklinikum, Mannheim, Germany, 3Gemeinschaftspraxis Hämatologie – Onkologie, Dresden, Germany, 4Dr. Aldaoud – Dr. Schwarzer Forschungsgesellschaft mbH, Leipzig, Germany, 5iOMEDICO, Freiburg i.Br., Germany, 6Praxis für Interdisziplinäre Onkologie & Hämatologie, Freiburg i.Br., Germany 1

5


CONTENTS AUTHOR INDEX

Conclusion: SIR biomarkers, routinely assessed in clinical settings can be easily used as prognostic and predictive markers to optimize therapeutic strategies in ABTC. To overcome the negative prognostic impact of high SIR combination chemotherapies should be recommended. Table 1: Association of SIR markers with OS

SIR marker

OS (months)

p-value

HR (95% CI), p-value

PLR >218 vs <218

10.6 vs 17.6

0.030

1.942 (1.056–3.576), 0.033

NLR >4.6 vs <4.6

9.4 vs 17.6

0.004

2.299 (1.273–4.154), 0.006

LMR <2.6 vs >2.6

13.8 vs 17.6

0.047

1.762 (0.999–3.110), 0.051

CRP >10mg/dl 3.8 vs 15.4 vs <10mg/dl

0.021

3.161 (1.126–8.873), 0.029

Immunotherapy of ovarian cancer

mGPS >2 vs <2

<0.001

47.951 (4.241–542.113), 0.002

1

0.7 vs 15.7

Disclosure: No conflict of interest disclosed. V28

H3K9-active demethylases disable oncogene-induced senescence, drive melanomagenesis, and are promising therapeutic targets Yu Y.1, Yue B.1, Ji S.2,3, Lohneis P.4, Werner-Klein M.5, Hummel M.4, Dörken B.1,2, Lee S.1,2, Schmitt C.1,2 Max-Delbrück-Center for Moleculare Medicine, Berlin, Germany, 2Charité – Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumor Immunology and Molekulares Krebsforschungszentrum, Berlin, Germany, 3Chinese Academy of Medical Sciences, Peking Union Medical College, Peking, China, 4Charité – Universitätsmedizin Berlin, Department of Pathology, Berlin, Germany, 5University Regensburg, Institute of Immunology, Regensburg, Germany 1

Introduction: Oncogene-induced senescence (OIS) is a failsafe program against imminent transformation and further propagation of (pre-)malignant cells. Paradigmatic are Ras- or BRAF-driven nevus cell nevi, whose senescent state serves as a barrier to melanoma formation. Mechanistically, histone H3 lysine 9-trimethylation (H3K9me3)-governed silencing of S-phase-promoting E2F target genes is essential for the sustained cell-cycle arrest. Given the importance of H3K9me3 and the frequent overexpression of H3K9-active demethylases in various cancer types, we hypothesized that candidate demethylases such as LSD1 and JMJD2C might exert their pro-tumorigenic potential via blocking OIS. Methods: Human and murine fibroblasts, primary human melanocytes, nevus and melanoma biopsies, melanoma cell lines and primary single disseminated melanoma cells (DMC) were subjected to functional analyses (expression, growth, senescence, transformation) in culture or as xenotransplants in nude mice after stable gene transfer or knockdown of relevant moieties, including LSD1, JMJD2C, oncogenic Ras and BRAF. In addition, pharmacological inhibition of LSD1 and JMJD2C was exploited in vitro and in vivo. Results: Ras- or BRAF-expressing mouse or human fibroblasts, as well as primary human melanocytes bypassed OIS if co-expressing LSD1 or JMJDC2. Moreover, stable expression of LSD1 or JMJD2C transformed Ras-driven mouse embryo fibroblasts. Importantly, activation of H3K9 demethylases in senescent cells enforced cell-cycle re-entry, i.e. licensed reversal of OIS (“escape”). On the contrary, inhibition of LSD1 or JMJD2C in demethylase- and Ras-overexpressing fibroblasts by RNA interference or chemical inhibitors restored senescence. We detected LSD1 and JMJD2C expression in melanoma biopsies, but not premalignant nevi samples, and found high-level H3K9-active demethylase expression as a prominent feature of melanoma cell lines. Inhibiting LSD1 or JM-

6

JD2C in melanoma cell lines or DMC induced cellular senescence, and controlled xenograft growth in nude mice. Remarkably, targeting LSD1 and JMJD2C even blocked melanoma progression of BRAF inhibitor (i.e. Vemurafenib)-resistant cells in vitro and in vivo. Conclusions: Our data unveil the essential but highly dynamic role of the H3K9me3 mark in OIS, the – in principle – intrinsic reversibility of this otherwise very stable condition, and suggest H3K9-active demethylases as promising targets for novel anti-cancer treatment strategies.

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed.

Fortbildung

Ovarial- und Uteruskarzinom V31

Weiss L.1, Mlineritsch B.1, Greil R.1 Paracelsus Medizinische Universität Salzburg, Universitätsklinik für Innere Medizin III, Salzburg, Austria

Ovarian cancer is the second most common gynecologic malignancy and with only 45% of patients surviving 5 years after diagnosis, new therapeutic strategies are urgently needed. A vast body of evidence suggests that in general ovarian cancer cells can be recognized by the immune system and the presence of tumor infiltrating T-cells is associated with a significantly better clinical outcome. Despite this immunogenicity overt disease can evolve and eventually patients may succumb to their disease – possibly due to immune evasion strategies exerted by the tumor. As one of these mechanisms, the Programmed Death-Ligand 1 (PD-L1) can be expressed by tumor cells and by binding to its receptor – the Programmed Death Protein 1 (PD-1) – it can down-regulate the function of T-cells. High expression of PD-L1 in ovarian cancer has been shown to be associated with significantly shorter survival. Earlier immunotherapy trials using intraperitoneal interleukin 2 and/or the adoptive transfer of tumor-infiltrating T-cells provided first evidence of clinical efficacy of immunotherapy in ovarian cancer. Also several vaccination studies could demonstrate an increase in tumor-specific T-cells which in some cases even conferred clinical benefit. Various tumor-associated antigens such as NY-ESO-1 or individual patients’ tumor cell lysate have been used for vaccination strategies. So called immune checkpoint inhibitors have shown unprecedented responses in notoriously difficult-to-treat cancers such as melanoma and lung cancer. The anti-PD-1 antibodies nivolumab and pembrolizumab, as well as the anti-PD-L1 antibodies BMS-936559 and avelumab are the first representatives of this new class of drugs having shown highly promising results in ovarian cancer, with some patients even achieving complete and also durable remissions. Combining different immunotherapeutic approaches might further increase the clinical efficacy in the future. Disclosure: Lukas Weiss: Other Financial Relationships: Reisekostenerstattung (Merck) Richard Greil: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag

V33

AML experimentell I

SH-2251 as a new possible treatment for AML

V32

Lams R.F.1, Botezatu L.1, Hönes J.1, Michel L.1, Köster R.1, Al-Matary Y.1, Dührsen U.1, Khandanpour C.1

DNMT3A mutations (DNMT3Amut) in acute myeloid leukemia (AML): monitoring of minimal residual disease (MRD). A study of the AML Study Group (AMLSG) Gaidzik V.I.1, Weber D.1, Paschka P.1, Krieger S.1, Kaumanns A.1, Krönke J.1, Kapp-Schwörer S.1, Köhne C.-H.2, Horst H.-A.3, Schmidt-Wolf I.G.H.4, Held G.5, Kündgen A.6, Ringhoffer M.7, Götze K.8, Kindler T.9, Fiedler W.10, Wattad M.11, Corbacioglu A.1, Bullinger L.1, Schlegelberger B.12, Thol F.12, Heuser M.12, Ganser A.12, Schlenk R.F.1, Döhner H.1, Döhner K.1 Uniklinik, Ulm, Germany, 2Klinikum, Oldenburg, Germany, 3Universitätsklinikum Schleswig-Holstein-Campus, Kiel, Germany, 4Universitätsklinikum, Bonn, Germany, 5Universitätsklinikum des Saarlandes, Homburg, Germany, 6 Universitätsklinikum, Düsseldorf, Germany, 7Städtisches Klinikum Karlsruhe GmbH, Karlsruhe, Germany, 8Klinikum rechts der Isar der Technischen Universität, München, Germany, 9Universitätsmedizin, Mainz, Germany, 10 Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany, 11Kliniken Essen Süd, Ev. Krankenhaus Essen-Werden gGmbH, Essen, Germany, 12 Medizinische Hochschule Hannover, Hannover, Germany 1

Introduction: The awareness of DNMT3Amut as a pre-leukemic lesion and founder mutation in AML as well as its presence in apparently healthy elderly brought new light into the disease. Within our treatment trials we sought to analyze if MRD monitoring in DNMT3Amut patients (pts) is clinically relevant and delivers further information for clonal hematopoiesis. Methods: We analyzed the DNMT3Amut R882H (n = 111) and R882C (n = 48) in AML pts entered on three AMLSG treatment trials [AML HD98A (n = 14; NCT00146120), AMLSG 07–04 (n = 87; NCT00151242), AMLSG 09–09 (n = 58; NCT00893399)] using a cDNA-based RQ-PCRassay by TaqMan technology (sensitivity between 10–3 and 10–4). Results: In total, 1,168 samples [bone marrow (BM), n = 615; peripheral blood (PB), n = 553] from 159 DNMT3Amut pts were analysed [diagnosis, n = 256; during therapy, n = 719; follow-up, n = 193]. Median BM DNMT3Amut transcript levels (TL) at diagnosis were 12690 (range, 0–54280); TL were not associated with known clinical characteristics, mutations in NPM1 or FLT3 or impact on OS and RFS. DNMT3Amut TL during therapy were significantly higher in BM than in PB after induction I, consolidation I and II (p = 0.01; p = 0.0003; p = 0.01). After double induction (DI) there was no difference in the median TL between 102 pts in complete remission (CR) and 12 pts not in CR. BM DNMT3Amut TL as log 10 transformed continuous variable during therapy had no influence on the endpoints death or relapse. The greatest TL reduction was seen after induction I (one log); after DI and end of therapy (ET), only 7/75 and 4/63 BM samples became MRD negative. At these two time-points MRD positivity did not impact OS (p = 0.89; p = 0.73) and remission duration (RD; p = 0.61; p = 0.30). Next we investigated the MRD DNMT3Amut log10-reduction (compared to level at diagnosis) with the median as a cut-off. There was no significant correlation for pts with a higher compared to a lower TL reduction for OS and RD after DI and ET (p = 0.87; p = 0.38; p = 0.67; p = 0.57). The BM DNMT3Amut TL as 4 increasing equally sized intervals according to the quartiles of the distribution revealed no prognostic impact after DI and ET on OS and RD (p = 0.24; p = 0.85; p = 0.38; p = 0.44). Conclusions: In our study neither DNMT3Amut TL nor its kinetics had a prognostic impact. Most pts showed persistent DNMT3Amut TL supporting the role of clonally derived hematopoiesis. By serial investigation of DNMT3Amut and its co-mutations the role of clonal evolution of AML will be further elucidated. Disclosure: No conflict of interest disclosed.

Abstracts

University Hospital Essen, Department of Hematology, Essen, Germany

1

Acute myeloid leukemia (AML) is a malignant neoplasia of the myelopoiesis. Despite intensive therapy the overall five year survival rate is under 30%, warranting new approaches. Oncorequisite factors like Growth factor independence 1 (Gfi-1) are promising new approaches to treat AML. Gfi-1 is a transcriptional repressor, which bind to and restricts p53 activity by modifying p53. In addition to this, it binds to the regulatory elements of pro-apoptotic genes such as Puma, Noxa (Pmaip1) and Bax (Bbc3). We could previously show that genetically inhibiting Gfi1 in leukemic cells leads to massive apoptosis in leukemic cells without side effects on normal cells. However no drugs are available to inhibit Gfi1 expression in patients. In this study, we examined the suitability of the novel thioamide-related compound SH-2251, which represses Gfi1 expression, to cure AML. To address this, we performed in vitro and in vivo studies using SH-2251, compound, which dramatically decreases Gfi1 mRNA expression. We treated different Gfi1 expressing human and murine AML cell lines such as Kasumi1, KG1, SKNO, HL-60 and C1498 by various concentrations of SH-2251. To rule out unspecific toxic effects we also testes non Gfi1 expressing cell lines. The results revealed that SH-2251 mediated inhibition of Gfi1 leads to an increased level of apoptosis and decreased cell numbers in the different Gfi1 expressing cell lines. Moreover, SH-2251 did not influence non-Gfi1 expressing tumor cell-lines, thus highlighting the specificity of this compound. To confirm our findings in vivo, we next investigated the effects of oral administration of SH-2251 (50mg/kg) in an AML mouse model. We transplanted mice with murine leukemic cells expressing the human oncofusion protein AML1/ETO9a, which can be recurrently found in AML patient cohorts. We observed that SH-2251 treatment inhibited AML development when compared with vehicle-administated control group. In summary, SH-2251 could be a new promising approach to treat Gfi1 expressing AML independent of their leukemia driving mutations, potentially opening a novel treatment approach for these patients. Disclosure: No conflict of interest disclosed. V34

Gfi1b plays an important role in initiation and progression of murine and human acute myeloid leukemia (AML) by regulating key oncogenic pathways Thivakaran A.1, Botezatu L.1, Hönes J.M.1, Zeller A.1, Michel L.1, Görgens A.2, Lennartz K.3, Köster R.1, Opalka B.1, Giebel B.2, Dührsen U.1, Khandanpour C.1 University Hospital Essen, Department of Hematology, Essen, Germany, University Hospital Essen, Institute for Transfusion Medicine, Essen, Germany, 3 University Hospital Essen, Institute for Cell Biology, Essen, Germany 1 2

Gfi1b (growth factor independence 1b) is a transcription factor important for the development of erythroid and megakaryocytic lineage. It also regulates quiescence and cell cycle progression of hematopoietic stem cells (HSCs) and early progenitor cells. We explored how different levels of Gfi1b influence the onset and progression of acute myeloid leukemia (AML). Published and own patient’s data indicated that Gfi1b is expressed at a lower level in leukemic blasts and leukemic stem cells (LSC) compared to non-malignant cells. To investigate the role of Gfi1b in initiation and progression of AML we used the Nup98/HoxD13 MDS mouse model that recapitulates key features of MDS including transformation to AML. To study the influence of different Gfi1b level on leukemia development, we have used two different mouse strains: Gfi1bGFP/WT mice, in which one allele of Gfi1b is replaced by GFP and a conditional mouse strain (Mx Cre tg Gfi1b fl/fl) in which the expression of Gfi1b can be abrogated after pIpC injection. We crossed these mice with Nup98HoxD13 MDS mice

Oncol Res Treat 2015;38(suppl 5):1–270

7


CONTENTS AUTHOR INDEX

and observed that Gfi1b heterozygosity accelerated AML development (p < 0.0001) compared to wt mice. Complete loss of Gfi1b resulted in a significantly earlier onset of AML (p = <0.0001). Loss of Gfi1b alone was not lethal for more than the 6 months. While all Gfi1b WT AML arising in the Nup98HoxD13 tg mice could be clearly attributed to the myeloid lineage, Gfi1b-deficient leukemic cells did not express any lineage marker but expressed very high levels of c-Kit, indicating that KO of Gfi1b is associated with immature leukemia. Gfi1b induces demethylation of H3K4 and deacetylation of H3K9. We found that Gfi1b deficient cells had a genomewide upregulation of H3K9 acetylation and methylation and that this was associated with pathways in stem cell and integrin regulation. Gfi1b regulates ITGB3 (integrin beta 3), which is required to promote growth and expansion of LSC. Conditional deletion of Gfi1b increased expression ITGB3 in bone marrow leukemic. On a molecular level, we found that loss of Gfi1b leads to increase levels of ROS (Reactive oxygen species). Excessive amounts of ROS causes the peroxidation of nucleic acids that could lead to carcinogenesis. In Gfi1b-KO leukemic cells gene expression arrays revealed upregulation of LSC genes and signaling prooncogenic pathways such as Wnt and hedgehog. Collectively, these data suggest that Gfi1b is a novel prognostic marker and tumor suppressor in AML. Disclosure: No conflict of interest disclosed. V35

Targeting aberrant NCAM (neural cell adhesion molecule; CD56) expression in acute myeloid leukemia Sasca D.1, Schüler A.1, Kriege O.1, Kunz K.1, Szybinski J.1, Fehr E.-M.1, Haehnel P.S.1, Gebhardt W.H.2, Reid G.2, Theobald M.1, Bullinger L.3, Kindler T.1 University Medical Center of Mainz, III. Medical Department, Hematology, Oncology, Pneumology, Mainz, Germany, 2Institute of Molecular Biology, Mainz, Germany, 3University Hospital of Ulm, Department of Internal Medicine III, Ulm, Germany 1

Introduction: Acute myeloid leukemia (AML) is a heterogeneous disease of the hematopoietic progenitor cell driven by the subsequent acquisition of genetic alterations. Approximately 20% of AML patients show strong expression of CD56 (neural cell adhesion molecule; NCAM), but the functional consequences have not been investigated to date. This study aims to examine the biological role of NCAM in AML and explore whether NCAM represents a potential therapeutic target. Methods: Leukemic cells were analyzed for proliferation, cell death, engraftment potential in xenografts and downstream signaling pathways after knock-down or overexpression of NCAM. Upstream regulatory mechanisms were investigated by DNaseI-hypersensitivity assays. To analyze NCAM effects at the stem cell level we used a well-established murine MLL-AF9 (MA9) leukemia model using C57BL/6 wild-type or NCAM/mice and performed serial replating and bone marrow (BM) transplantation experiments. Results: NCAM was highly expressed in several cell lines and correlated with MA9+ leukemia. Knockdown of NCAM caused diminished proliferation, G1-arrest and finally cell death. Suppression of NCAM sensitized leukemic blasts to genotoxic stress in vitro and prolonged survival of xenografts, whereas exogenous overexpression in NCAM-negative cell lines caused an increased resistance to treatment. Applying DNaseI-hypersensitivity assays we demonstrate accessible binding sites for the transcription factors MEIS1, MEF2c and STAT1. shRNA-mediated knockdown of MEIS1, MEF2c and MLL-AF9 resulted in downregulation of NCAM expression, suggesting an upstream regulatory role for MLL-AF9. Analysis of downstream signaling pathways upon knockdown of NCAM revealed reduced expression levels of beta-Catenin and its downstream products but also activation of SMAD2/3, upregulation of p21 and downregulation of BCL2. To analyze NCAM expression in leukemia-initiating cells (LICs), we transduced wild-type and NCAM-/- derived BM cells with MA9 expressing retrovirus and transplanted syngeneic recipient mice. LICs (Lin-c-Kit+CD34+FcgR+) showed strong surface expression of NCAM,

8

Oncol Res Treat 2015;38(suppl 5):1–270

whereas normal HSCs (Lin-c-Kit+Sca1+) were NCAM-negative. Recipients of NCAM-/-MA9_cells developed leukemia with prolonged latency, and in colony assays replating was diminished. Conclusions: Our data suggest that NCAM is involved in self-renewal and stress resistance of leukemic cells. Targeting of NCAM could become a promising therapeutic strategy in AML. Disclosure: No conflict of interest disclosed. V36

Targeting of acute myeloid leukemia initiating cells with Fc optimized CD96 antibodies Staudinger M.1, Peipp M.1, Kellner C.1, Bulduk M.1, Humpe A.1, Gramatzki M.1 Universität Kiel, Sektion f. Stammzell- & Immuntherapie, 2. Med. Klinik, Kiel, Germany 1

Introduction: Acute myeloid leukemia (AML) is maintained by a population of leukemic stem cells (LSC). LSC residing in patients after intensive chemotherapy account for the high rate of relapse observed in AML patients. The development of novel approaches for efficient elimination of LSC is a prerequisite to improve AML therapy. Previously we demonstrated with our antibody TH-111 that CD96, a surface antigen identified on AML-LSC, represents a suitable target for ex vivo purging of autologous stem cell grafts. Since certain situations would require antibody application in the patient, here, a novel Fc optimized antibody against CD96 was designed to allow targeting of AML-LSC in vivo. Methods: CD96 antibodies with Fc variants optimized for antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) were generated. Effector mechanisms triggered by the antibody variants were analyzed. NK cells from healthy donors were used as effector cells. Plasma served as source of complement. ADCC and CDC activity against cell lines and freshly isolated tumor cells was analyzed by flow cytometry or 51Cr assays. Colony forming assays were performed to analyze the cytotoxic potential of the novel CD96 antibody variants against hematopoietic progenitor cells (HPC). Results: The variable regions of an affinity maturated scFv were used for generating full length CD96 antibodies with engineered IgG1-based Fc variants optimized for ADCC or CDC activity. ADCC was identified as the major effector mechanism. The ADCC-optimized CD96 antibody efficiently recruited NK cells to lyse KG1a myeloid blasts as well as primary AML tumor cells. The cytotoxic activity was compared to a similarly designed antibody directed against CD34, to secure that this ADCC-optimized CD96 antibody discriminates LSC from healthy HPC. Both, CD34+ HPC from healthy donors and CD34+/CD96+ KG1a cells were lysed by the CD34 antibody. In contrast, our CD96 antibody did not kill CD34+ HPC but efficiently eliminated KG1a. The lack of stem cell toxicity was supported by colony forming assays demonstrating that the capacity of HPC to proliferate and differentiate was not affected. Conclusion: The particular features of Fc-optimized CD96 antibodies lacking stem cell cytotoxicity but eliminating AML-LSC may provide an additional therapeutic option for AML patients in certain clinical situations for instance in the setting of allogeneic stem cell transplantation. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V37

Targeted resequencing of MLL-PTD positive AML patients reveals a high prevalence of co-occurring mutations in epigenetic regulator genes Herold S.1,2, Stange T.3, Kuhn M.3, Platzbecker U.1, Roeder I.3, Röllig C.1, Serve H.4, Berdel W.E.5, Bornhäuser M.1, Ehninger G.1, Thiede C.1 Universitätsklinikum Dresden, Medizinische Klinik I, Dresden, Germany, Deutsches Krebsforschungszentrum, Deutsches Konsortium für Translationale Krebsforschung DKTK Dresden, Heidelberg, Germany, 3TU Dresden, Institut für Medizinische Informatik und Biometrie, Dresden, Germany, 4 Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt, Germany, 5 Medizinische Klinik A, Medizinische Fakultät Münster, Münster, Germany 1

Disclosure: Sylvia Herold: Expert Testimony: Illumina Christian Thiede: Stock Ownership: Ownership Agendix; Expert Testimony: Illumina.

Freier Vortrag

Multiples Myelom klinisch

2

Introduction: Partial tandem duplications within the MLL gene (MLLPTD) are a recurrent molecular alteration in AML (up to 10%) and are associated with a poor prognosis. Little is known on the molecular profile and cooperating mutations in patients with MLL-PTD. In order to identify accompanying mutations, we performed whole exome sequencing (WES) of 8 MLL-PTD patients. Based on the observed alterations we then resequenced 118 MLL-PTD patients using either a custom targeted panel or a comprehensive commercial amplicon panel. Methods: Patients included in this analysis were treated in prospective protocols of the Study Alliance Leukemia (SAL). We performed WES of FACS-sorted leukemic blasts and T-cells. For targeted resequencing, in total 1407 amplicons (custom panel) and 568 amplicons (TruSight Myeloid panel) were sequenced. Results: WES of 8 MLL-PTD patients revealed a total number of 490 SNVs. Most frequently mutated genes were the epigenetic regulators DNMT3A, IDH1/2 and TET2. Confirmed somatic mutations were also found in genes less known to be mutated in AML, such as ATM, GNAS, TET1 and EP300. The targeted resequencing of 118 patients revealed a total number of 249 individual mutations, 16 genes were not found to be mutated, and in 8 patients, no co-occurring mutations were identified. Most frequently mutated genes in the entire MLL-PTD cohort were DNMT3A (33.9%), FLT3 (33.1%; 27.9% ITD; 5.0% TKD), IDH1 (22.9%), IDH2 (28.8%) and TET2 (19.5%) (Fig.1), whereas typical NPM1 mutations were rarely seen (2.4%). Especially mutations in IDH1, IDH2, and TET2 were found very frequently and about twice as common as in the general AML patient population. Conclusions: Our analysis did not reveal any new and specific individual aberration present in patients with MLL-PTD mutations. However, our data indicate a very high prevalence of alterations in epigenetic regulator genes (close to 80%), which strongly argues for a particular disease biology in MLL-PTD patients and might also implicate that treatment based on demethylating agents or histone-deacetylase inhibitors might be especially attractive for patients with MLL-PTD.

Fig. 1. Percentage of mutated genes.

Abstracts

V38

Single versus tandem high-dose melphalan followed by autologous blood stem cell transplantation in newlydiagnosed multiple myeloma: final results from the GMMGHD2 trial Mai E.K.1, Benner A.2, Bertsch U.1, Schmidt-Wolf I.G.H.3, Hänel A.4, Kunzmann V.5, Naumann R.6, Neben K.7, Egerer G.1, Hillengass J.1, Neubauer A.8, Peyn A.9, Ko Y.-D.10, Peter N.11, Salwender H.J.12, Scheid C.13, Goldschmidt H.1,14, German-speaking Myeloma Multicenter Group (GMMG) University Clinic Heidelberg, Department of Internal Medicine V, Heidelberg, Germany, 2German Cancer Research Center (DKFZ), Division of Biostatistics, Heidelberg, Germany, 3University Clinic Bonn, Center for Integrated Oncology, Medizinische Klinik und Poliklinik III, Bonn, Germany, 4Klinikum Chemnitz gGmbH, Department of Internal Medicine III, Chemnitz, Germany, 5 University Clinic Würzburg, Department of Internal Medicine II, Würzburg, Germany, 6Gemeinschaftsklinikum Mittelrhein, Department of Hematology and Oncology, Koblenz, Germany, 7Klinikum Baden Baden, Department of Hematology and Oncology, Baden Baden, Germany, 8University Clinic Giessen/ Marburg, Department of Hematology and Oncology, Marburg, Germany, 9 Klinikum Bremen Mitte, Department of Internal Medicine I, Bremen, Germany, 10 Evangelische Kliniken Bonn, Department of Hematology and Oncology, Bonn, Germany, 11Carl-Thiem Klinikum Cottbus, Department of Internal Medicine II, Cottbus, Germany, 12Asklepios Hospital Hamburg Altona, Department of Hematology and Oncology, Hamburg, Germany, 13University Hospital Köln, Department of Internal Medicine I, Köln, Germany, 14Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg, Germany 1

Introduction: The role of upfront single versus tandem high-dose melphalan (HDM) followed by autologous blood stem cell transplantation (ASCT) in multiple myeloma (MM) remains controversial. The open-label, randomized, multicenter phase III trial HD2 is the only prospective trial that applied the recommended HDM of 200mg/m2 prior to ASCT in the single and tandem group. Methods: Between 12/1998 and 07/2002, 385 patients were randomized to receive either single (arm A) or tandem (arm B) HDM+ASCT after VAD/VID/VCAP-like induction therapy (IT). Main inclusion criteria were Salmon/Durie stage II/III, age ≥18 or <66 years (at initial diagnosis) and at least stable disease (SD) after IT. The primary analysis of the study compared event-free survival (EFS) two years after randomization. Results: The intention-to-treat (ITT) population consisted of 177/181 (arm A/B) patients, respectively. In arm B, 47 patients refused second HDM+ASCT due to non-medical reasons. Non-inferiority of single HDM+ASCT versus tandem HDM+ASCT regarding 2-year EFS was demonstrated (ITT, HR = 0.93 with a one-sided confidence limit of 0.77). A per-protocol (PP) analysis including patients who received the assigned treatment (single/tandem HDM+ASCT, n = 156/n = 93 patients) revealed a non-significant trend towards prolonged EFS in the tandem HDM+ASCT group (p = 0.12). The CR rates before/after first HDM+ASCT were comparable in both groups. In arm B, CR rates significantly improved after second HDM+ASCT (ITT, after first/second HDM+ASCT: 15.1% vs. 19.4%, p = 0.04). Achieving a CR before and after first/second HDM+ASCT significantly improved EFS (p = 0.02) and OS (p = 0.03) in the whole study population. On long-term follow-up no OS differences were observed (ITT/PP: p = 0.33/0.90). Conclusions: We observed a trend towards a prolonged EFS in arm B in the PP population, which did not translate into the ITT population. With a median follow-up of more than 11 years, no OS differences were observed. The high number of patients refusing second HDM+ASCT confound these findings. Tandem HDM+ASCT significantly increased

Oncol Res Treat 2015;38(suppl 5):1–270

9


CONTENTS AUTHOR INDEX

CR rates, and achieving a CR remained an important prognostic factor to prolong EFS and OS in the whole study population. Therefore, tandem HDM+ASCT can constitute an important therapeutic approach to improve CR rates. Subgroups that benefit from tandem HDM+ASCT and the role of single versus tandem HDM+ASCT remain to be defined in the era of novel agents. Disclosure: Elias Mai: Other Financial Relationships: Travel grants from Janssen-Cilag, Celgene, Onyx, and Mundipharma, outside the submitted work Hartmut Goldschmidt: Advisory Role: Personal fees and other support from Janssen-Cilag, Celgene, Novartis, Onyx, Millennium, Chugai, outside the submitted work; Financing of Scientific Research: Personal fees and other support from Janssen-Cilag, Celgene, Novartis, Onyx, Millennium, Chugai, outside the submitted work; Expert Testimony: Grants from Janssen-Cilag, Celgene, Novartis, Chugai, outside the submitted work. V39

IKZF1 expression is associated with outcome in lenalidomide treated newly diagnosed multiple myeloma Kuchenbauer F.1, Krönke J.1, Kull M.1, Teleanu V.1, Bullinger L.1, Straka C.2, Mügge L.-O.3, Bassermann F.4, Engelhardt M.5, Bargou R.6, Einsele H.6, Knop S.6, Langer C.1, Deutsche Studiengruppe Multiples Myelom (DSMM) Universitätsklinik Ulm, Innere Medizin III, Ulm, Germany, 2Hematology and Oncology, Schön Klinik Starnberger See, Berg, Germany, 3Friedrich Schiller University, Internal Medicine II, Jena, Germany, 4Klinikum Rechts der Isar, Department of Medicine III, München, Germany, 5University of Freiburg, Hematology and Oncology, Freiburg, Germany, 6University Hospital of Würzburg, Department of Internal Medicine II, Würzburg, Germany 1

Introduction: Lenalidomide is an immunomodulatory drug (IMiD) with high activity in multiple myeloma (MM). IMiD binding to the CRBN E3 ubiquitin ligase results in targeted ubiquitination and degradation of the lymphoid transcription factors IKZF1 and IKZF3 and constitutes a unique mechanism of action for a class of drugs. Methods: We analyzed mRNA expression levels of IKZF1, IKZF3, and CRBN by real-time quantitative PCR (RQ-PCR) in CD138-selected pre-treatment samples from 60 pts with newly diagnosed MM uniformly treated within a phase II clinical trial of the German Myeloma Study Group (DSMM). Additionally, all pts were characterized for the presence of cytogenetic abnormalities using a comprehensive set of FISH probes. All patients received induction consisting of four cycles of lenalidomide, adriamycin and dexamethasone followed by high-dose melphalan (Mel200) with autologous stem cell transplantation (aSCT). Genetically defined low- and intermediate risk patients received a second aSCT, while those pts with high-risk cytogenetics (presence of a del17p13, t(4;14) or t(14;16)) underwent allogeneic stem cell transplantation if a donor was available. All patients received lenalidomide maintenance for 12 months. Results: Median expression levels normalized to plasma cells from healthy volunteers of CRBN, IKZF1, and IKZF3 were 0.66 (range, 0.12–2.86), 0.66 (0.16–2.92), and 0.52 (0.19–5.13), respectively. Expression levels of CRBN, IKZF1, and IKZF3 were not affected by the presence of chromosomal aberrations (del13q14, del17p13, del9q34, +1q21, t(4;14), t(11;14), t(14;16)). Patients achieving a complete response (CR) or very good partial response (VGPR) had a trend towards a lower IKZF1 expression than patients achieving a partial response (PR) (median IKZF1 expression: 0.62 vs 0.84, p = 0.07). Consistently, lower IKZF1 expression levels were associated with a better outcome: when segregating IKZF1 expression levels into quartiles, pts with the lowest (Q1) quartile of IKZF1 expression had a 3 year progression-free survival (PFS) of 86% compared to 51% in patients of the remaining quartiles (Q2-Q4)(Log rank test, p = 0.01). This translated into a better overall survival (OS) (IKZF1 Q1:100% vs IKZF1 Q2–4:74% after 3 years, log rank p = 0.03). In contrast, CRBN and IKZF3 expression levels had no impact on response, PFS, or OS. Disclosure: Florian Kuchenbauer: No conflict of interest disclosed. Christian Langer: Advisory Role: Celgene, Janssen-Cilag, Bristol-Myers Squibb.

10

Oncol Res Treat 2015;38(suppl 5):1–270

V40

Comparative assessment of myeloma response to induction treatment in the GMMG MM5 study using IMWG criteria and hevylite assay Scheid C.1, Hose D.2, Bertsch U.3, Hielscher T.4, Kunz C.4, Salwender H.5, Haenel M.6, Merz M.2, Mai E.K.2, Schurich B.2, Munder M.7, Schmidt-Wolf I.8, Gerecke C.9, Lindemann W.10, Zeis M.11, Weisel K.12, Dürig J.13, Jauch A.14, Peters-Regehr T.15, Zorn M.16, Goldschmidt H.3, GMMG Klinik I für Innere Medizin, Uniklinik, Köln, Germany, 2Medizinische Klinik V, Universitätsklinikum, Heidelberg, Germany, 3Sektion Multiples Myelom der Medizinischen Klinik V und des Nationalen Centrums für Tumorerkrankungen (NCT), Universität Heidelberg, Heidelberg, Germany, 4Biostatistics, German Cancer Research Center, Heidelberg, Germany, 5Asklepios Hospital Altona, Hamburg, Germany, 6Klinikum Chemnitz gGmbH, Chemnitz, Germany, 7 University Medical Center, Mainz, Germany, 8Center for Integrated Oncology (CIO) and Medizinische Klinik und Poliklinik III, University of Bonn, Bonn, Germany, 9Hematology and Oncology, Helios-Hospital Berlin Buch, Berlin, Germany, 10Kath. Krankenhaus, Hagen, Germany, 11Department of Hematology and Stem Cell Transplantation, Asklepios Hospital St. Georg, Hamburg, Germany, 12Dept. of Hematology, Oncology and Immunology, University of Tuebingen, Tübingen, Germany, 13University Hospital, Essen, Germany, 14 Institute of Human Genetics, University Hospital, Heidelberg, Germany, 15The binding site GmbH, Schwetzingen, Germany, 16Sektion Klinische Chemie, Universitätsklinikum, Heidelberg, Germany 1

Introduction: Treatment of multiple myeloma has substantially improved over the last decade. However response assessment still relies on parameters such as serum protein electropheresis or immunofixation. A new response category of stringent CR was introduced using information on serum free light chains and bone marrow clonality. Recently the hevylite assay measuring kappa and lambda-restricted IgG and IgA was introduced. The objective of this study was to analyse the value of using the hevylite assay for assessing ealry MM response. Methods: The MM5 study of the German GMMG randomised 604 newly diagnosed myeloma-patients to receive bortezomib, dexamethasone and either adriamycin or cyclophosphamide as induction before high-dose chemotherapy. Response after 3 cycles of induction was assessed using IMWG criteria. Clonal IgG and IgA was measured by hevylite in frozen serum samples from study baseline and after induction in 154 patients. Results: 109 patients had IgG-paraprotein with 84 (77%) kappa and 25 (23%) lambda while 45 patients had IgA with 34 (76%) kappa and 11 (24%) lambda. Response to induction according to IMWG criteria was CR in 8 (5.2%), nCR in 19 (12.3%), VGPR in 27 (17.5%), PR in 71 (46.1%), MR in 19 (12.3%), SD in 7 (4.5%) and missing in 3 patients (1.9%). No patient had sCR based on free-light chain ratios. The frequencies of normal hevylite kappa/lambda ratios in the different IMWG response categories were as follows: IgG-myeloma: CR :1/2 (50%), nCR: 2/4 (33.3%), VGPR: 1/20 (4.8%), PR: 3/52 (5.5%), MR: 0/16, SD: 1/4 (20%), missing: 0/2. IgA- myeloma: CR: 4/6 (66.7%), nCR: 2/13 (15.4%), VGPR: 2/6 (33.3%), PR: 3/15 (20%), MR: 0/3, SD: 0/2, missing: 0/0. Persistent pair suppression of non-clonal Ig was found in 85/109 (78%) for IgG and 30/46 (65%) for IgA. Conclusion: Triplet induction achieved a CR/nCR rate of nearly 20%, while hevylite kappa/lambda normalisation occurred in 7.5% (IgG) and 24% (IgA). There was little overlap to IMWG response. This may be related to the fact that hevylite response not merely mirrors paraprotein response, but also captures suppression of the non-involved pair. It seems warranted to validate hevylite response after induction treatment with progression-free and overall survival once further follow-up data becomes available. Disclosure: Christof Scheid: Financing of Scientific Research: binding site, Janssen, Celgene, Amgen Hartmut Goldschmidt: Expert Testimony: binding site, Janssen, Celgene, Bristol Myers Squibb.

Abstracts


CONTENTS AUTHOR INDEX

V41

Molecular profiling in patients with plasma cell myeloma treated with novel agents Medinger M.1, Jakab A.1, Halter J.1, Heim D.1, Buser A.1, Gerull S.1, Stern M.1, Passweg J.1 Universitätsspital Basel, Hämatologie, Basel, Switzerland

1

Introduction: Angiogenesis plays an important role in the pathogenesis and in progression of plasma cell myeloma (PCM). Novel agents like thalidomide, lenalidomide or bortezomib have in part anti-angiogenic properties. In this study we examined gene expression of angiogenic molecules in patients with PCM and correlated these markers to treatment response to novel agents. Methods: We included 103 PCM patients treated with novel agents IMiDs (thalidomide or lenalidomide)-based regimens, (bortezomib (V)-based regimens or a combination of IMiD and bortezomib-based regimen (IMiD/V). Total RNA from peripheral blood mononuclear cells was isolated before anti-myeloma therapy and after 3 cycles. The mRNA levels of angiogenic molecules were measured using the Human Angiogenesis RT2 Profiler PCR Array. Changes in gene expression between responders and non-responders were analyzed as a fold increase/decrease. The response evaluation was performed after 3 cycles. Results: Of the 103 patients included 32 patients were treated with an IMiD-based regimen, 34 with a V-based regimen and 37 with the combination (IMiD/V). The CR + VGPR rate was 43% in the IMiD-based group, 51% in the V-based group and 55% in the combination group (IMiD/V). Baseline gene expression of 11 from 84 genes tested correlated with advanced disease stage (p < 0.005 in each case). Regarding all 103 patients, gene expression of 15 from 84 genes tested (pre and post-treatment and changes in levels pre-treatment/post-treatment) were significantly different in responders compared to non-responders. Responders had a decreased expression proangiogenic factors (e.g., IL-6, VEGFA, ANGPT2, NRP1, FGFR, ECGF, and MMP-9) and increased expression of antiangiogenic factors (e.g., TIMP-3, TGF-β, and CXCL10). In the different treatment groups, the angiogenic gene levels (14/84) were significantly different in responders compared to non-responders in the IMiD-based group and the combination group after 3 cycles of therapy but not in the V-based group. Conclusions: In the IMiD-containing therapy groups we found significant changes of angiogenic gene expression in responders compared to non-responders, whereas in the bortezomib-based group the difference in angiogenic gene expression was not significant. Possibly, anti-angiogenic properties play a higher role in IMID based treatment as compared to proteasome inhibitor based treatment. Disclosure: No conflict of interest disclosed. V42

Vorinostat (V), bortezomib (B), doxorubicin (Dox) and dexamethasone (Dex, VBDD) in relapsed or refractory multiple myeloma patients (pts): results of an open, non-comparative, phase I/II investigator initiated trial (IIT) Keller A.1,2, Waldschmidt J.M.1,2, Wider D.1, Jakobs D.1, Möller M.1, Reinhardt H.1, Pantic M.1, Grishina O.1, Ihorst G.3, May A.4, Frey A.4, Kohlweyer U.1, Jung M.5, Duyster J.1,2, Wäsch R.1,2, Engelhardt M.1,2 Freiburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 2CCCF, Freiburg, Germany, 3 Freiburg University Medical Center, Early Clinical Trials Unit (ECTU), Freiburg, Germany, 4Freiburg University Medical Center, Department of Pathology, Freiburg, Germany, 5Freiburg University, Institute of Pharmaceutical Sciences, Freiburg, Germany 1

Introduction: Vorinostat (V) was chosen as an epigenetic agent within this VBDD-regimen, since it has shown promising anti-tumor effects as a histone deacetylase inhibitor (HDACi). V inhibits the enzyme activity of HDAC1, 2, 3 and 6, allowing the activation of tumor suppressor genes, thereby slowing down cancer cells. Synergistic effects of V and B have

12

Oncol Res Treat 2015;38(suppl 5):1–270

been shown. V antagonizes escape mechanisms secondary to B-based therapies. Dox and Dex were chosen in this VBDD-regimen, because they are active and approved in relapsed and refractory (RR) MM. Since MM relapse leading to RRMM is common, novel treatment combinations are vitally needed. The aim of this phase I/II IIT was to test the tolerability and activity of the VBDD schedule within an out-pt regimen for RRMM pts. Methods: V was escalated from 100mg (dose level 0), to 200mg (+1) and 300mg (+2). Primary objectives (MTD; 3+3 dose escalation), secondary objectives (safety, IMWG/EBMT responses, PFS, OS) and correlative endpoints (organ function, prognostic factors, QoL, comorbidity and HDAC-activity in PBMNCs/BM) were assessed throughout the trial. Dose limiting toxicities (DLTs) were defined as any possibly drug-related adverse event (AEs) ≥grade 3 (CTCAE) during the 1st cycle. Results: 33 RRMM pts have been enrolled (median age 63 (47–78), KPS 90% (70–100%). The number of prior therapy lines was substantial with a median of 3 (1–8; with prior B, SCT and IMiDs in 88%, 94% and 42%, respectively). No DLTs have been observed. SAEs amounted to 15 and occurred in 9/33 pts (27%): amongst them 2 nonfatal SAEs judged to be related to the investigational therapy (1 bacteraemia, 1 herpes zoster reactivation). The response was rewarding with best ORR (>PR) and clinical benefit rate (CBR; >SD) of 62% and 97%, and end of treatment (EoT) ORREoT (>PR) and CBREoT (>SD) of 45% and 83%, respectively. Preliminary data propose that the response was independent of unfavorable cytogenetics. Comorbidity assessment assured no decline in pts´ condition. Ongoing pharmacodynamic analyses show HDAC downregulation after the 2nd cycle. Conclusions: VBDD is an effective and well-tolerated out-pt regimen with promising response rates in heavily pretreated RRMM pts. The employed VBDD-regimen, with a continuous, rather than pulsed V-schedule, constitutes a promising treatment option, expands current standard therapies and, similarly to other HDACi, suggests V as a valuable add-on within this combination schedule. * equal contribution AK + JW. Disclosure: Alexander Keller: No conflict of interest disclosed. Monika Engelhardt: Financing of Scientific Research: Ja; Immaterial Conflict of Interests: Educational grants by MSD, Janssen-Cilag, Comprehensive Cancer Center Freiburg (CCCF) and Biotera fundation (ME) V43

Successful treatment of patients with newly diagnosed/ untreated light chain multiple myeloma with a combination of bendamustine; prednisone and bortezomib (BPV) Pönisch W.1, Mrachacz H.2, Khoder N.3, Plötze M.3, Holzvogt B.3, Andrea M.3, Schliwa T.3, Heyn S.3, Pfrepper C.4, Franke G.N.3, Krahl R.3, Jentzsch M.3, Leiblein S.3, Schwind S.3, Vucinic V.3, Niederwieser D.3, OSHO, Ostdeutsche Studiengruppe für Hämatologie und Onkologie Universitätsklinikum Leipzig, Hämatologie, internistische Onkologie, Leipzig, Germany, 2Universitätsklinikum Leipzig, Hämatologie, Leipzig, Germany, 3 Universitätsklinikum, Leipzig, Germany, 4Diakonissenkrankenhaus, Leipzig, Germany 1

Background: Patients with light chain myeloma have frequently a light chain tubular cast nephropathy, which can lead to severe renal impairment. Objective: Both bortezomib and bendamustine have been identified as quickly acting, effective and well tolerated drugs and might therefore constitute an adequate combination regimen for patients with newly diagnosed/untreated light chain multiple myeloma. Methods: Between September 2009 and March 2014, 20 patients with newly diagnosed/untreated light chain multiple myeloma were treated with bendamustine 60 mg/qm on days 1 and 2, bortezomib 1.3 mg/qm on days 1, 4, 8 and 11, and prednisone 100 mg on days 1, 2, 4, 8 and 11 once every 21 days. 5 patients (25%) had a moderate or severe renal dysfunction (eGFR 15–59 ml/min) and 9 patients (45%) a renal failure/dialysis (eGFR < 15 ml/min).

Abstracts


CONTENTS AUTHOR INDEX

Results: The median number of the BPV-treatment was 2 (1–5) cycles. 19 patients (95%) responded after at least one cycle of chemotherapy with 3 sCR, 4 nCR, 5 VGPR, and 7 PR. The myeloma light chains decreased rapidly, reaching the best response after the first cycle in 8 and after the second cycle in additional 9 patients. 16 patients discontinued therapy after median 2 cycles of BPV treatment to receive autologous (n = 13) or autologous/allogeneic SCT (n = 3). All together 10/14 patients with at least moderate renal failure improved their renal function (4 CRrenal, 2 PRrenal, 4 MRrenal). 3 of the 6 dialysis-dependent patients became dialysis-independent. With a median follow up of 23 months of the surviving patients, median PFS and OS for patients at 24 months were 90% and 95%, respectively. The most common severe side effect was grade 3–4 leukocytopenia in 25% of the patients. Grade 3–4 thrombocytopenia was observed in 15% of the patients. Moderate to severe infection were seen in 4 patients. Conclusion: We conclude that BPV is effective and well tolerated in patients with newly diagnosed/untreated light chain multiple myeloma. Disclosure: No conflict of interest disclosed.

Freier Vortrag Lungenkarzinom I V44

Tumor-infiltrating B lymphocytes characterized by CD79a and MUM1 independently predict outcome in patients with nonsmall cell lung cancer Fischer R.N.1, Scheel A.H.2, Rothschild S.I.1,3,4, Schlößer H.A.3,5, Wolf J.1, Büttner R.2, Ansén S.1, von Bergwelt-Baildon M.S.3 Uniklinik Köln, Centrum für Integrierte Onkologie (CIO), Lung Cancer Group Cologne, Köln, Germany, 2Uniklinik Köln, Institut für Pathologie, Köln, Germany, 3 Uniklinik Köln, Klinik I für Innere Medizin, Cologne Interventional Immunology, Köln, Germany, 4Universitätsspital Basel, Medizinische Onkologie, Basel, Switzerland, 5Uniklinik Köln, Klinik und Poliklinik für Allgemein-, Viszeral- und Tumorchirurgie, Köln, Germany 1

Introduction: Tumor-infiltrating lymphocytes play an important role in cell-mediated immune-destruction of cancer cells and tumor growth control. T-cell infiltrates are robust prognostic biomarkers in a variety of solid tumors. In non-small cell lung cancer (NSCLC) a prognostic role of T cell subtypes, natural killer cells and dendritic cells within the tumor stroma has been described. Here, we studied the role of tumor-infiltrating B cells characterized by CD79a (B-cell antigen receptor complex-associated protein alpha chain) and MUM1 surface expression (Multiple myeloma oncogene 1) in patients with NSCLC. To our knowledge, this study represents the so far largest cohort analyzing the prognostic impact of tumor-infiltrating B cells in NSCLC. Methods: B cell infiltration was quantified using immunohistochemistry and antibodies to CD79a (Dako, clone JCB117) and MUM1 (Dako, clone MUM1p) on tissue microarrays of paraffin embedded tumor sections. Genetic driver mutations were identified by next-generation sequencing and FISH analysis. Results: 478 tissue samples from NSCLC patients were available for final analysis. Median age of the patient population was 66 years with 65% male patients. 56% had adenocarcinoma and 39% squamous cell histology. 65% of patients were diagnosed with localized disease (stage I: 44%; stage II: 21%), 30% locally advanced disease (stage III) and 6% were diagnosed with stage IV. CD79a and MUM1 positive cells were detected in 40.8% (195/478) and 40.2% (192/478) of the analyzed NSCLC tissue samples, respectively. B cell infiltration was not associated with clinical or histo-pathological characteristics. CD79a expression was associated with a trend towards a better outcome (median OS 49 vs. 40 months, p = 0.069). The expression of MUM1 showed a significantly prolonged overall survival (median OS 54 vs. 40 months, p = 0.025). In the multivariate analysis B cell infiltration characterized by positivity for CD79a and MUM1 was an independent prog-

Abstracts

nostic marker for survival (p = 0.045) as was MUM1 expression alone (p = 0.031). Conclusions: In this cohort of patients with localized and locally advanced stage NSCLC, B cell infiltration characterized by immunohistochemical positivity for CD79a and MUM1 represents an independent prognostic marker. This finding supports the hypothesis of a B cell-mediated anti-tumor immunity. Disclosure: No conflict of interest disclosed. V45

Do we miss changes in quality of life? Data from the LuLife project in patients with non-small cell lung cancer in Germany von Verschuer U.1, Sandner R.2, Däßler K.-U.3, Tessen H.W.4, Münz M.5, Spring L.5, Jänicke M.5, Marschner N.6 Hämato-Onkologische Gemeinschaftspraxis, Essen, Germany, 2Passauer Onkologische Praxis Dres. Prenninger & Sandner, Passau, Germany, 3 Onkologische Schwerpunktpraxis, Freital, Germany, 4Onkologische Schwerpunktpraxis, Goslar, Germany, 5iOMEDICO, Freiburg, Germany, 6Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany 1

Introduction: Most patients (pts) with lung cancer are diagnosed with advanced disease and receive palliative chemotherapy. Both tumor burden and chemotherapy can strongly affect quality of life (QoL) but little is known about how QoL changes over time in pts in routine care. The LuLife project prospectively investigated qol and other patient reported outcomes (PROs) in real-life pts. Methods: LuLife was conducted with the tumor registry on lung cancer (TLK), which prospectively collects data on routine treatment of lung cancer as administered by office-based medical oncologists in Germany and was started in 2010. In LuLife the validated questionnaires EORTC QLQ-C30 (global QoL) and -LC13 (lung cancer module) were sent to pts with NSCLC at start of 1st-line treatment and then every eight weeks up to six times, and filled at home. Here, data on global as well as cancer- and symptom-related QoL over time are presented. Results: 507 pts answered at least one questionnaire. They had a mean age of 66 years at the start of 1st-line treatment. ECOG status was 0/1/≥2 for 30/56/14% of pts. Most pts (>80%) received platin based chemotherapy. Questionnaires were answered by 485/424/353/290/240/194 pts at 0/8/16/24/32/40 weeks which represents a return rate of 96/87/81/76/73/65% of all pts alive at the respective time points. In total 38% of the pts returned the last questionnaire. Overall, the better the PRO values at baseline, the more questionnaires were returned. Analysis of mean difference to baseline revealed no mean change in global QoL, pain, fatigue and emotional function over time, but small to medium clinically relevant mean deterioration in social, physical, role and cognitive function. Alopecia strongly increased within the first two months, while dyspnoe (small change) and neuropathic symptoms (large change) continuously worsened over time. Despite little mean changes from baseline in the whole population, analysis of the time to deterioration revealed that, after 16 weeks 30% of all pts had reported clinically relevant decreased QoL (individual deterioration of at least 10% of the score) and worsened pain, but far more worsened fatigue (50%) or dyspnoe (40%). Conclusion: Our data show that PROs of NSCLC pts change over time but results, especially on global QoLQoL, differ depending on the method of choice. It is crucial to select appropriate analyses to avoid missing changes in PROs, which could indicate a need to improve patient care. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

13


CONTENTS AUTHOR INDEX

V46

V47

Randomized study of individualized, pharmacokineticallyguided dosing of paclitaxel combined with carboplatin in advanced non-small cell lung cancer patients

P53 disruptive mutation is a negative predictive factor in EGFR M+ NSCLC treated with TKI

Joerger M.1, Salamone S.J.2, von Pawel J.3, Kraff S.4, Fischer J.R.5, Eberhardt W.E.6, Gauler T.C.7, Müller L.8, Reinmuth N.9, Reck M.9, Kimmich M.10, Mayer F.11, Kopp H.-G.11, Behringer D.M.12, Ko Y.-D.13, Früh M.1, Hilger R.A.7, Roessler M.14, Moritz B.14, Miller M.C.2, Jaehde U.4, CESAR Central European Society for Anticancer Drug Research-EWIV Cantonal Hospital, St. Gallen, Switzerland, 2Saladax Biomedical Inc., Bethlehem, United States, 3Asklepios Fachkliniken, Klinik für Pneumologie, Gauting, Germany, 4Institute of Pharmacy, Clinical Pharmacy, University, Bonn, Germany, 5 Klinik, Löwenstein, Germany, 6West German Cancer Center, Ruhrlandklinik, Essen, Germany, 7Universitätsklinikum, Essen, Germany, 8Onkologische Schwerpunktpraxis, Leer, Germany, 9LungenClinic, Großhansdorf, Germany, 10 Klinik Schillerhöhe, Gerlingen, Germany, 11Universitätsklinikum, Tübingen, Germany, 12Augusta-Kranken-Anstalt, Bochum, Germany, 13Johanniter Krankenhaus, Bonn, Germany, 14CESAR Central Office, Wien, Austria 1

Introduction: Variability of chemotherapy exposure may cause severe toxicity or lack of efficacy. Paclitaxel (PTX) exposure (time above a plasma concentration of 0.05 mM, Tc > 0.05) has been shown to predict toxicity. Whereas carboplatin dosing is adapted to kidney function, PTX dosing only accounts for body-surface area. We developed a PTX dosing algorithm for avoidance of supra- or subtherapeutic PTX exposure based on Tc>0.05 determined from a single blood sample drawn 18–30 hours after starting PTX infusion. This study was initiated to validate the use of a pharmacokinetically (PK)-guided dosing strategy for PTX to optimize exposure in patients with advanced non-small cell lung cancer (NSCLC). Methods: 304 advanced NSCLC patients were randomly assigned to receive up to 6 cycles of first-line 3-weekly carboplatin AUC6 combined with PTX either at a standard dose of 200 mg/m2 (Arm A) or at a PK-guided dose (Arm B). Initial PTX dose in Arm B was between 150 to 200 mg/ m2 based on age and sex. PTX exposure was determined from a single blood sample drawn 18–30 hr after infusion initiation using an HPLC-UV assay. PTX doses were adjusted according to the previous cycle exposure to target a Tc>0.05 between 26 and 31 hours using our dosing algorithm. Primary objective of the study was to detect an 11% reduction of grade 4 neutropenia with PK-guided PTX dosing. Secondary objectives included comparison of neuropathy rates, tumor response and progression-free survival (PFS) between the study arms. Results: Compared to standard dosing, PK-guided dosing of PTX reduced the incidence of grade 4 neutropenia (measured on day 15 of each cycle) (21% vs. 15%, P = 0.029), grade ≥2 neuropathy (27% vs. 14%, P < 0.001), and grade ≥3 neuropathy (8% vs. 1%, P < 0.001). PK-guided dosing of PTX led to a dose reduction in 62% and a dose increase in 17% of the patients. The proportion above the target exposure was reduced from 41% in cycle 1 to 2% in cycle 6. Objective response rate between Arms A and B was similar (32% vs. 29%, P = 0.70), as was the median PFS (5.2 vs. 4.7 months, hazard ratio 1.1, 95% CI 0.8–1.4, P = 0.54). Conclusion: PK-guided dosing of PTX improved the risk-benefit profile in patients with advanced NSCLC. The dosing algorithim resulted in reduction of grade 4 neutropenia and neuropathy ≥ grade 3, without reducing efficacy. Disclosure: No conflict of interest disclosed.

Lüers A.C.1,2, Neemann N.1, Prenzel R.3, Scriba D.C.4, Willborn K.C.5, Stropiep U.2, Falk M.6, Hallas C.6, Tiemann M.6, Griesinger F.1,2 Pius-Hospital, University Department Internal Medicine-Oncology, Oldenburg, Germany, 2Pius-Hospital, Hematology and Oncology, Oldenburg, Germany, 3 Pius-Hospital, Pneumology, Oldenburg, Germany, 4Pius-Hospital, Thoracic Surgery, Oldenburg, Germany, 5Pius-Hospital, Radiotherapy, Oldenburg, Germany, 6Hematopathology, Hamburg, Germany 1

Introduction: p53 mutations are common in lung cancer, and have also been described in EGFR mutated patients. The impact of p53 mutations in EGFR M+ patients is controversial, especially if classified as “disruptive” and “non-disruptive” according to their functional effect on the p53 protein as proposed by Poeta and colleagues. The aim of the study was therefore to systematically analyze EGFR and p53 mutations within a cohort of patients with lung cancer stage IV (UICC 7), to correlate alterations with clinical characteristics and to investigate a potential impact of p53 mutations on treatment outcome. Methods: 267 patients from a single center diagnosed with lung cancer stage IV were studied for the presence of EGFR as well as inactivating p53 mutations. Methods for the detection of EGFR mutations included Sanger Sequencing and hybridization based COBAS testing. P53 mutations were detected by Sanger Sequencing. Clinical characteristics including smoking status were available for all patients. Results: 267 consecutive patients at the lung cancer center of the Pius-Hospital Oldenburg were studied. The overall EGFR mutation rate was 19% (51/267) in all patients, 80% (41/51) showing common mutations of exon 19 or 21. P53 disruptive mutation showed in 16% (8/51) and p53 nondisruptive mutation occurred in 11% (22/51) whereas p53 WT was found in 47% (24/51). In 8/51 (16%) patients p53 analysis was not successful. OS was 37 months in p53 disruptive mutation and p53 WT patients compared to 19 months in p53 nondisruptive mutation (p < 0,05). PFS on 1st line TKI therapy was 18 months in p53 nondisruptive mutation and p53 WT patients and 6 months in p53 disruptive mutation (p < 0,024). Similar results could be shown in the EGFR common mutation subgroup but not in the uncommon mutation subgroup. Conclusion: Significant differences in PFS and OS in EGFR M+ patients were observed depending on p53 mutation status. P53 mutational status is only predictive when disruptive and non-disruptive P53 mutations are differentiated. P53 should be tested prospectively in EGFR M+ patients as management of patients on 1st line TKI may be different. Disclosure: Anne Lüers: Advisory Role: Boehringer Ingelheim; Expert Testimony: Astra Zeneca Frank Griesinger: Advisory Role: Boehringer Ingelheim, Roche, Pfizer, Astra, Novartis, Lilly, MSD, BMS, Clovis; Expert Testimony: Astra Zeneca. V48

Evaluation of disease-related symptoms in patients (pts) with advanced squamous (SQ) non-small cell lung cancer (NSCLC) treated with nivolumab (NIVO) or docetaxel (DOC) Gralla R.J.1, Coon C.2, Taylor F.2, Penrod J.R.3, DeRosa M.2, Dastani H.3, Orsini L.3, Reck M.4 Albert Einstein College of Medicine, Bronx, United States, 2Adelphi Values, Boston, United States, 3Bristol-Myers Squibb, Princeton, United States, 4 Lungenklinik Großhansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany 1

Background: CheckMate 017 (NCT01642004), a randomized, open-label, global phase 3 study, evaluated efficacy and safety of 2nd-line NIVO vs DOC in advanced SQ NSCLC pts. Overall survival was significantly superior and duration of treatment longer for NIVO vs DOC. Disease-related symptoms were also evaluated using the Lung Cancer Symptom Scale (LCSS), with scores reported from 0 (least severe) to 100 (most severe). Methods: Assessment was performed every 4 weeks (Q4W) for NIVO and Q3W for DOC for the first 6 mos on treatment, then Q6W for the rest of

14

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

the treatment period (both arms), and at follow-up visits 1 and 2 (post discontinuation). At each assessment, mean baseline (BL) and mean change from BL of the LCSS average symptom burden index (ASBI) were summarized by arm. Secondary endpoint was proportion of pts with clinically meaningful improvement in LCSS ASBI by wk 12 (≥10 point decrease, the minimally important difference [MID]) based on all randomized pts. Results: Pt BL characteristics were generally balanced across treatment arms. LCSS completion rates at BL and ≥1 subsequent assessment were 68.9% for NIVO and 62.8% for DOC. Completion rates remained relatively consistent throughout assessments and by treatment arm. BL LCSS ASBI values were similar for NIVO (29.6; SD 16.4) and DOC (29.6; SD 14.7). By wk 12, 20.0% (27/135; 95% CI 13.6, 27.7) of NIVO vs 21.9% (30/137; 95% CI 15.3, 29.8) of DOC pts demonstrated clinically meaningful symptom improvement. The NIVO arm had statistically significant mean improvements from BL at each LCSS ASBI assessment from wks 12–54, after which sample sizes dropped to < 10 pts; mean improvement from wks 40–54 exceeded MID. DOC pts remaining on treatment had no statistically significant changes in LCSS ASBI through wk 18, after which the sample dropped to < 10 pts. At follow-up visits 1 and 2, mean LCSS ASBI for both NIVO and DOC pts indicated similar worsening of symptoms relative to BL (range, 5.5–9.5); for DOC pts, differences from BL were statistically significant. Conclusion: The proportion of pts with meaningful symptom improvement by wk 12 was similar for both NIVO and DOC, but overall average symptom burden on NIVO improved from BL over most of the available follow-up period. Average symptom burden for DOC pts remained stable relative to BL during their shorter time on treatment. The results show statistically and clinically significant symptom reductions from BL for 2nd-line SQ NSCLC pts treated with NIVO. Disclosure: Richard Gralla: Advisory Role: Bristol-Myers Squibb Martin Reck: Advisory Role: Hoffman La-Roche, Lilly, Merck Sharpe & Dohme, Bristol-Myers Squibb, AstraZeneca, Boehringer-Ingelheim, Pfizer, Novartis. V49

further oncogene mutation could be detected. In 8 patients (14.0%) no other additional genetic aberrations were found. Conclusion: High-level cMET amplification can occur with and without additional genetic lesions. About one third of high-level MET-amplified patients had a co-occurring KRAS mutation. In order to detect patients with high-level cMET amplification, it is not sufficient to concentrate on EGFR-mutated patients in the resistance setting. Further work is needed if these findings translate into clinical consequence. Disclosure: Anna Eisert: No conflict of interest disclosed. Jürgen Wolf: Advisory Role: Advisory boards (compensated) and lecture fees: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Expert Testimony: Research support: Bayer, Boehringer-Ingelheim, Novartis, Pfizer, Roche.

Wissenschaftliches Symposium Fortgeschrittenes Prostatakarzinom V52

Biomarker in mCRPC Machtens S.1 Marienkrankenhaus, Zentrum für Innere Medizin, Urologie und Kinderurologie, Leverkusen, Germany 1

Advances in the therapy of metastatic castration-refractory prostate cancer make an individual approach neccessary. This individualised decision can be supported by new biomarker. These biomarker can have an prognostic, predictive or monitoring character. An overview is delivered in regard to current developments in the evaluation of new biomarkers and their possible influence on Treatment stratifications. New aspects concerning the splice variants of the androgen receptor, gene fusions and others are highlighted.

Clinical and genetic presentation of high-level cMET-amplified Non-small cell lung cancer (NSCLC) with adenocarcinoma histology

Disclosure: Stefan Machtens: Financing of Scientific Research: Amgen, Sanofi Aventis, Novartis, Astellas, Janssen.

Eisert A.1, Scheffler M.1, Gogl L.1, Frank R.1, Scheel A.1, Merkelbach-Bruse S.1, Michels S.1, Fischer R.1, Büttner R.1, Wolf J.1

Fortbildung

Uniklinik, Köln, Germany

1

Introduction: cMET (MNNG HOS Transforming gene) is a proto-oncogene encoding the hepatocyte growth factor receptor (HGFR) with HGF as the only known ligand. Whereas the role of cMET mutations in NSCLC patients remains debated controversively, amplification of cMET is considered as one of the molecular resistance mechanisms in EGFR-mutated NSCLCs treated with 1st or 2nd generation EGFR-TKIs. Further, at least for high-level amplifications, these aberrations might be a possible target for cMET-targeted therapy also in EGFR-wt patients. Patients and methods: Beside the screening of rebiopsied EGFR-TKI-resistant patients, cMET amplification status as assessed by fluorescence in-situ hybridization (FISH) was also analyzed in all presenting NSCLC patients with adenocarcinoma histology, regardless of therapy line or stage. A cohort of 1599 patients within a regional screening network was analyzed using FISH and next-generation sequencing (NGS). High-level amplifications were specified by the following criteria: cMET/CEN7 ratio of ≥ 2 or an average cMET gene copy number per cell of ≥ 6 or ≥ 10% of tumor cells containing ≥ cMET signals (according to Schildhaus et al., Clinical Cancer Research 2014). Results: In a time period of one year 57 patients with high-level cMET amplification were identified: 35 patients (61.4%) were male and 22 patients (38.6%) were female. Of 41 patients analyzed for stage so far, 27 (66.9%) had metastatic disease (stage IV). cMET amplifications co-occurred with other driver mutations, especially EGFR (n = 7, 12.3%) and KRAS (n = 18, 31.6%). In 24 patients (42.1%) mutations within TP53 without

16

Oncol Res Treat 2015;38(suppl 5):1–270

Supportivtherapie bei neuen systemischen Therapieformen V66

Endocrine side effects of anticancer drugs Capraro J.1 Kantonsspital Aarau, Endokrinologie, Diabetologie, Metabolismus, Aarau, Switzerland 1

Several of the newer anticancer drugs have been associated with distinct endocrine adverse effects. Regular screening and a high level of suspicion may help to prevent significant morbidity. Immunotherapies against immune checkpoints that inhibit T cell activation (cytotoxic T lymphocyte antigen 4 (CTLA-4)) and programmed cell death 1 (PD-1) have been found to adversely affect several endocrine tissues with hypopituitarism and adrenal failure being the most severe sequelae. Thyroid dysfunction may be another important associated condition. Central hypothyroidism is a well-recognized side effect of bexarotene. Older immunomodulating drugs, such as interferon-a and interleukin-2 are known to induce variably high incidence of autoimmune thyroid dysfunction. Tyrosine Kinase Inhibitors (TKI’s) often induce thyroid disorders and regular testing of thyroid function is mandatory. Therapy with mTOR inhibitors may result in severe dyslipidemia and diabetes mellitus. TKI’s on the other hand may be associated with improved glucose metabolism in patients with diabetes, or with frank hypoglycemia. Abiraterone acetate, through its distinct effect on steroid metabo-

Abstracts


CONTENTS AUTHOR INDEX

lism, leads to adrenal mineralocorticoid excess which must be prevented by concomitant glucocorticoid administration. Oncologists must be aware of these side effects in order to provide regular screening and initiate a specific diagnostic workup if suspicious clinical signs are present. If hormonal changes are detected early referral to an endocrinologist and multidisciplinary management are key to successful therapy. Disclosure: No conflict of interest disclosed.

Fortbildung

Nierenzellkarzinom – neue Aspekte und Standards V67

Management of side effects and adherence to oral cancer therapies in metastatic renal cell cancer Rothermundt C.1 Kantonsspital St. Gallen, Onkologie/Hämatologie, St. Gallen, Switzerland

1

Owing to targeted therapies and sequencing treatments, the life expectancy of patients with metastatic renal cell cancer (mRCC) has been extended from 13 months in the cytokine era (Motzer 2002) to over 30 months (Motzer 2014) today. All available drugs for mRCC are long-term therapies with continuous or intermittent dosing, which are given until disease progression or intolerable toxicity occurs. Thus patients spend long times “on treatment” (Negrier 2012). Retrospective data show that only 52%–79% of mRCC patients receive second-line treatment, depending on the choice of first-line treatment (Levy 2013), and 13% of patients receive third-line treatment (Iacovelli 2013). Tolerability of therapies and side effect management is therefore of importance in mRCC. Maintenance of an effective dose is a concern in patients treated with oral targeted therapies, in order to achieve optimal results. Decisions on dose reductions and interruptions should always be made knowing about potential concessions on efficacy. There are several comprehensive reviews on side effects and side effect management in patients on targeted therapies for mRCC (Lacouture 2008, Kollmannsberger 2011, Eisen 2012, Mendez-Vidal 2012, Larkin 2014), concerning e.g. cardiovascular, cutaneous, constitutional, endocrine, gastrointestinal, laboratory and organ function toxicity. Progress in cancer treatment has been accompanied by a paradigm shift in medication delivery: a shift in treatment away from the outpatient clinic and into the patient’s home. There has been little emphasis on making sure patients take medications correctly once prescribed—the final step in the bench to patient pathway. However, the success of oncologic treatment depends on the patient’s adherence to medication regimens (Gellad 2014). Factors associated with nonadherence include treatment regimen, the patient, social environment, and the clinician-patient interaction. Patient perceptions and motivations appear to be the most important determinants. Prediction of nonadherence is especially challenging. Interventions that have produced some improvement in adherence among oncologic patients include educational programs; behavioral modification techniques, and use of reminder systems and cues (Partridge 2002). In Switzerland an educational program for oncology nurses and medical oncologists is underway to raise awareness for adherence, to establish adherence assessments and to plan interventions aimed at improving adherence. Disclosure: Christian Rothermundt: Advisory Role: Pfizer; Financing of Scientific Research: GSK, Novartis.

Abstracts

V68

Checkpoint inhibitors – a new road to salvation in mRCC? Grünwald V.1 MHH, Hannover, Germany

1

Checkpoint inhibitors have enriched the armentarium to treat cancer in recent years. Its implementation as the fifth cornerstone of cancer treatment is still novel and we will have to explore its role in the field in more detail in the upcoming years in order to understand its full scope. It is the start of a new era, with many research questions ahead. Inhibitors of the programmed death receptor 1 (PD-1) or its ligand (PDL1) have unleashed the T-cell response, through enhancing the cytotoxic T-cell response. Early clinical studies in metastatic renal cell carcinoma (mRCC) confirmed an objective response rate (ORR) of 20–22%, which warrants further drug development in this disease. More interestingly, PD-1 inhibition achieved a promising overall survival (OS) of 18–25 mo., which raises expectations in previously treated patients. Therefore a current phase III study tested PD-1 inhibition against everolimus in later lines of therapy, and results are awaited later this year. CTLA4 is another T-cell checkpoint in control of the T-cell priming phase. The combination of CTLA4 and PD-1 inhibition is thought to enhance T-cell priming, while boosting cytotoxic T-cell response. Early clinical studies suggested an increase in ORR to 43–48% and render this approach a candidate for future drug development whether as adjuvant or aggressive palliative treatment with the aim to improve survival. However, much more checkpoints are known to control T-cell fate, and novel agents are being developed to target them. Results are eagerly awaited and may forge a complete new landscape in mRCC, were improvement in OS may be reached. It is a very encouraging time in oncology, with great expectations ahead of us. Disclosure: Viktor Grünwald: Advisory Role: BMS, MSD, Pfizer, Novartis, GSK, Bayer; Financing of Scientific Research: BMS, Pfizer, Novartis, GSK.

Freier Vortrag

Lymphome aggressiv klinisch I V71

MRD eradication should be the therapeutic goal in mantle cell lymphoma and may enable tailored treatment approaches: Results of the intergroup trials of the European MCL Network Pott C.1, Macintyre E.2, Delfau-Larue M.-H.3, Ribrag V.4, Unterhalt M.5, Kneba M.6, Hiddemann W.5, Hermine O.7, Dreyling M.5, Hoster E.5, EU-MCL study group University Hospital Schleswig-Holstein, Second Department of Medicine, Kiel, Germany, 2Laboratoire d’Hématologie, Hôpital Necker-Enfants-Malades, Paris, France, 3Institut Mondor de Récherche Biomédicale (IMRB), Département d’Immunologie, Dérmatologie et Oncologie, Créteil, France, 4Laboratoire de récherche translationelle, Institut Gustave Roussy, Villejuif, France, 5Department of Internal Medicine III, University of Munich, Großhadern, Germany, 6Second Medical Department, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany, 7Department of Clinical Hematology, University Paris Descartes, Necker Hospital, Paris, France 1

Introduction: Minimal residual disease (MRD) for response assessment allows prediction of the clinical course and prognosis prior to clinical study endpoints in B-cell malignancies. Based on the randomized intergroup trials of the EU-MCL Network for stage II-IV mantle cell lymphoma (MCL) investigating immuno-chemotherapy followed by autologous stem cell transplantation (ASCT) for patients <65 yrs and rituximab or interferon maintenance for patients >65 yrs, we prospectively monitored MRD. Methods: MRD was analysed in peripheral blood (PB) after induction and in 3-monthly intervals after ASCT or 2-monthly during maintenance until progression. Patients in complete or partial remission 6 months after ASCT/end of induction (MCL Younger/MCL Elderly) were included. MRD status was defined as negative, if there were only nega-

Oncol Res Treat 2015;38(suppl 5):1–270

17


CONTENTS AUTHOR INDEX

tive MRD values, or positive in case of at least one positive MRD value during a 6-months period. Landmark analyses were performed for progression-free (PFS) and overall survival (OS) according to MRD status for each 6-months period until 4 years of follow-up. Results: Among patients in remission 6 months after ASCT/end of induction, MRD results during follow-up were available in 406 patients (255 MCL Younger,151 MCL Elderly). The distribution into low, intermediate, and high risk MIPI of 44%, 34% and 22%, respectively, was similar to the total study population. During follow-up, about 20–25% of patients were MRD+ during the first 3 years, and less than 20% in the subsequent follow-up. In each landmark analysis, a positive MRD status was highly associated with a shorter PFS and OS. This association was independent of baseline MIPI score, treatment arm or protocol. Thus, the prognostic value of MRD positivity was maintained in both trials MCL Younger and MCL Elderly. Remarkably, landmark analyses at all time points indicated a strong association also of low-level MRD with a shortened PFS. Finally, MRD analyses in 76 patients at relapse confirmed the close association of clinical relapse to MRD positivity, with only 10 patients (13%) being PB-negative. MRD reappearance preceeded clinical relapse in median 18 months. Conclusion: Our data demonstrate that achievement and preservation of MRD response is the strongest independent predictor of prognosis in patients with MCL. Applying MRD status as tool for tailored treatment, therapeutic approaches should focus on a maximum MRD response to improve long term outcome. Disclosure: No conflict of interest disclosed. V72

Acute hepatitis E infection in a young adult patient receiving rituximab for Burkitt lymphoma Matschke J.1, Alashkar F.1, Gerken G.2, Dührsen U.1, Jochum C.2 Department of Hematology, University Hospital Essen, University of DuisburgEssen, Essen, Germany, 2Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany 1

Introduction: Monoclonal antibodies like anti-CD20/rituximab have a high relevance for the therapy of B-cell-lymphoma. The risk for reactivation of hepatitis B was often described. Hepatitis E virus infections are rare and mostly self-limiting. Acute hepatitis E infections during a rituximab containing chemotherapy are not well described so far. Case presentation: A 21 year old patient was diagnosed in November 2013 with Burkitt lymphoma. The chemotherapy regimen was chosen according to the GMALL-B-ALL/NHL 2002 study of the German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia (GMALL) and was administered between November 2013 and June 2014. Three different courses (A,B,C) of chemotherapy were given in a 3-week interval and each was once repeated. Rituximab was given with the standard dose of 375 mg/m2 at the beginning of each course and with two additive cycles at the end of the six courses for a total of 8 administrations. The chemotherapy was well tolerated without major complications. After the fifth course of chemotherapy increasing transaminases were noticed (AST 109 U/L, ALT 123 U/L), which increased in the following two months (AST 414 U/L, ALT 739 U/L). Bilirubin concentrations were normal. A duplex sonography of the liver was without pathologic findings. Serological diagnostics excluded hepatitis A-C,E and cytomegalovirus. Two months after first observation of elevated transaminases and after the 8th administration of Rituximab a HEV viral load was detected using PCR. The patient developed a chronic hepatitis E during the following months. Seven months after the last course of rituximab B-cells reconstituted (CD19+-cells: 150/ nl). Two weeks after reconstitution of B-cells hepatitis E PCR in blood and stool was negative and transaminases were normalized. Conclusion: Patients with unknown acute or chronic hepatitis should be tested for hepatitis E. Acute hepatitis E infections could become chronically during immunosuppression, especially after administration of rituximab. B-cell reconstitution is important for curing hepatitis E.

V73

Long-term safety and efficacy of single-agent Ibrutinib in patients with relapsed or refractory mantle cell lymphoma: Updated results of an international, multicenter, open-label phase 2 study Dreyling M.1, Wang M.L.2, Rule S.3, Martin P.4, Goy A.5, Auer R.6, Kahl B.S.7, Jurczak W.8, Advani R.H.9, Romaguera J.E.2, Williams M.E.10, Barrientos J.C.11, Chmielowska E.12, Radford J.13, Stilgenbauer S.14, Jedrzejczak W.W.15, Johnson P.16, Spurgeon S.E.17, Zhang L.2, Baher L.18, Cheng M.18, Beaupre D.M.18, Blum K.A.19 University Hospital LMU, Munich, Germany, 2The University of Texas MD Anderson Cancer Center, Houston, United States, 3Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, United Kingdom, 4 Weill Cornell Medical College, New York, United States, 5John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, United States, 6Barts Health National Health Service (NHS) Trust, London, United Kingdom, 7University of Wisconsin School of Medicine and Public Health, Madison, United States, 8Jagiellonian University, Krakow, Poland, 9Stanford University School of Medical College, New York, United States, 10University of Virginia School of Medicine, Charlottesville, United States, 11Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, United States, 12 Oddzial Kliniczny Onkologii, Centrum Onkologii, Bydgoszcz, Poland, 13The University of Manchester and the Christie NHS Foundation Trust, Manchester, United Kingdom, 14Universitatsklinikum Ulm, Klinik fur Innere Medizin II, Ulm, Germany, 15Medical University of Warsaw, Warsaw, Poland, 16Cancer Research UK Clinical Centre, Southampton General Hospital, Southampton, United Kingdom, 17Oregon Health and Science University, Portland, United States, 18Pharmacyclics, Inc., Sunnyvale, United States, 19Ohio State University Comprehensive Cancer Center, Columbus, United States 1

Introduction: Ibrutinib is a first-in-class, oral inhibitor of Bruton’s tyrosine kinase approved for MCL patients who have received at least 1 prior therapy. Previous results of the international, multicenter, open-label phase 2 trial demonstrated the promising activity and acceptable safety profile of ibrutinib in relapsed or refractory (R/R) MCL (Wang et al., N Engl J Med. 2013). Here, we present the updated safety and efficacy results of this trial at a median 26.7-month follow-up. Methods: Patients (N = 111) received oral ibrutinib 560 mg once daily, and those with stable disease or better could enter a long-term extension study. The primary end point was investigator-assessed overall response rate (ORR) per the 2007 revised IWG criteria. Adverse events (AEs) were characterized by preferred terms using MedDRA version 16.1 and were evaluated over 6-month time intervals (1–6, 7–12, 13–18, 19–24, >24 months). Results: The median patient age was 68 years (range, 40–84), with median 3 prior therapies (range, 1–5) and prior bortezomib in 43%. The median treatment duration was 8.3 months; 46% of patients were treated for >1 year, and 22% were treated for ≥2 years. The ORR was 67% (23% complete response) with a median duration of response of 17.5 months. The 24-month PFS and OS rates were 31% (95% CI: 22.3–40.4) and 47% (95% CI: 37.1–56.9), respectively. The most common AEs in >30% patients included diarrhea (54% all grade, 5% grade ≥3), fatigue (50% all grade, 5% grade ≥3), nausea (33% all grade, 1% grade ≥3), and dyspnea (32% all grade, 5% grade ≥3). All-grade infections were reported in 78% of patients, with the most frequent grade ≥3 infections being pneumonia (8%), urinary tract infection (4%), and cellulitis (3%). Any-grade bleeding was experienced by 50% of the patients, and grade ≥3 bleeding events in ≥2% patients were hematuria (2%) and subdural hematoma (2%). Common all-grade hematologic AEs were thrombocytopenia (22%), neutropenia (19%), and anemia (18%). Treatment discontinuation due to AEs was reported in 11% of patients. The prevalence of infection, diarrhea, and bleeding was highest for the first 6 months of therapy and less thereafter. Conclusions: Ibrutinib continues to demonstrate a favorable benefit/risk profile in R/R MCL. Approximately one-third of patients remain progression-free at 2 years. The additional follow-up further supports the safety and durability of response with ibrutinib in MCL and does not reveal an increase in unforeseen AEs.

Disclosure: No conflict of interest disclosed.

18

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Disclosure: Martin Dreyling: Advisory Role: Pharmacyclics; Financing of Scientific Research: Pharmacyclics Kristie Blum: Expert Testimony: Pharmacyclics/Janssen. V74

Analysis of clinical characteristics and prognosis of patients with previously untreated diffuse large B-cell lymphoma of the gastrointestinal tract Lehners N.1, Krämer I.1, Koschny R.2, Herfarth K.3, Egerer G.1, Ho A.D.1, Witzens-Harig M.1 Uniklinik Heidelberg, Medizinische Klinik V, Heidelberg, Germany, 2Uniklinik Heidelberg, Medizinische Klinik IV, Heidelberg, Germany, 3Uniklinik Heidelberg, Radiologische Klinik, Heidelberg, Germany 1

Introduction: The gastrointestinal tract is one of the more frequent sites of extranodal involvement of diffuse large B-cell lymphoma (DLBCL). However, gastrointestinal DLBCL represents a very heterogeneous disease encompassing various biological entities. Therefore, optimal therapeutic strategies remain a matter of debate. Methods: Patients with newly diagnosed DLBCL and gastrointestinal involvement presenting to the University Hospital of Heidelberg between 01/2000 and 12/2011 were included in this analysis. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. The impact of variables on PFS and OS was evaluated by univariate log-rank tests as well as by multivariate Cox regression. Results: 82 patients were identified, 47 were male, 35 female. Median age was 60.5 years [range 19–86 years]. Site of involvement was stomach in 37 (45%), duodenum in 4 (5%), small intestine in 19 (23%), colon in 8 (10%) and multiple gastrointestinal sites in 14 (17%). 24 patients (29%) had IPI 0–1, 27 (33%) IPI 2–3, and 13 (16%) IPI 4–5; 18 patients had no evaluable IPI score. 5-year PFS was 65%, 5-year OS was 79%. There was no significant difference in regard to survival between the different sites of gastrointestinal involvement both in uni- and multivariate analysis. While neither age, sex, stage, and IPI score had a significant impact on survival in multivariate analysis, presence of B-symptoms was significantly associated with inferior PFS (p = 0.02) yet not with OS. In regard to therapy, the addition of rituximab did not show a significant impact in regard to survival both in uni- and multivariate analysis. However, escalation of chemotherapy from CHOP to a more aggressive regimen (in our cohort mainly CHOEP) had significant positive impact on PFS (p = 0.004) and OS (p = 0.04) in univariate analysis; in multivariate analysis, it was significantly associated with prolonged PFS (p = 0.03) and showed a trend towards prolonged OS (p = 0.09). Patients receiving CHOEP had an excellent outcome with 5-year PFS 90% and 5-year OS 100%. While consolidative radiotherapy did not have an impact in univariate analysis, there was a trend towards beneficial impact on PFS (p = 0.09) and OS (p = 0.06) in multivariate analysis. Conclusions: In our cohort of patients with DLBCL and gastrointestinal involvement escalation of chemotherapy to a more aggressive regimen than CHOP as well as consolidative radiotherapy seem to be beneficial therapeutic strategies. Disclosure: No conflict of interest disclosed.

V75

1st-line treatment of patients with high-grade non-Hodgkin’s lymphoma: Similar effectiveness with R-CHOP-14 or R-CHOP-21 – Data from the German prospective TLN Registry Knauf W.1, Abenhardt W.2, Mohm J.3, Rauh J.4, Grugel R.5, Harde J.5, Jänicke M.5, Marschner N.6, TLN Registergruppe Onkologische Gemeinschaftspraxis, Frankfurt a.M., Germany, 2Münchner Onkologische Praxis Elisenhof, München, Germany, 3Onkologische Gemeinschaftspraxis, Dresden, Germany, 4Fachinternistische Gemeinschaftspraxis u. Therapiezentrum, Witten, Germany, 5iOMEDICO, Freiburg i. Br., Germany, 6Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg i. Br., Germany 1

Introduction: R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone) is the standard of care for patients (pts) with previously untreated high-grade (aggressive) non-Hodgkin’s lymphoma (aNHL). Dose intensification of CHOP has shown ambiguous results. Whether the dose-dense two-weekly schedule (R-CHOP-14) is superior to the three-weekly schedule (R-CHOP-21) is a matter of debate. While clinical trials are restricted to highly selected pts, clinical registries offer insight into effectiveness of real-life treatment. Methods: The open prospective clinical registry on lymphoid neoplasms (TLN Registry, ClinicalTrial.gov NCT00889798) documents the treatment of pts with lymphoid B-cell neoplasms by German office-based haematologists. Pts are followed for 5 years (yrs). Data regarding pts and tumour characteristics, co-morbidities, all systemic treatments and outcome are recorded. Between May 2009 and August 2014, a total of 3,798 pts have been recruited by 122 study sites. Results: Of 565 pts with aNHL recruited at the start of 1st-line therapy with R-CHOP, 41% were treated with R-CHOP-14 and 59% received R-CHOP-21, with a tendency in favour of R-CHOP-21 over time. Pts were 68 yrs old, 46% female, 25% presented with tumour stage IV (Ann Arbor) and 64% with at least one co-morbidity. Pts characteristics showed no considerable differences between the two schedules. CHOP was applied for 6 cycles (median), rituximab for 8 cycles in R-CHOP-14 and 6 cycles in R-CHOP-21 (median). G-CSF was given in 98% (R-CHOP-14) and 63% (R-CHOP-21) of pts. Objective response rate (ORR) as assessed by the local site was 96% (R-CHOP-14) and 93% (R-CHOP-21); the clinical (unconfirmed) complete remission rate was 65% (R-CHOP-14) and 66% (R-CHOP-21). With a median follow-up of 25 months, 2yr progression-free survival rate (PFSREG) is 77% (R-CHOP-14) and 86% (R-CHOP-21). 2yr overall survival rate (OS) is 87% (R-CHOP-14) and 86% (R-CHOP-21). 10% of pts have received 2nd-line therapy. Overall, 10% of pts have been lost to follow-up. At this point, the high rate of pts alive without progression (>80%) precludes multivariate regression analyses regarding factors affecting PFS or OS. Conclusion: Our data show that in routine practice in Germany, pts with previously untreated high-grade NHL and treated with R-CHOP are more likely to receive the three-weekly schedule R-CHOP-21. First outcome data show that the effectiveness (ORR, PFSREG and OS) of both schedules is similar. Disclosure: No conflict of interest disclosed.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

19


CONTENTS AUTHOR INDEX

Fortbildung

Gesundheitspersonal neue Rollen, neue Aufgaben

Freier Vortrag MPN II / CML II

V76

V80

Shaping careers of the future

A new monoclonal antibody (CAL2) detects all types of CALRETICULIN mutations in formalin-fixed and paraffin embedded bone marrow biopsies

Baumgartner U.1 Kalaidos Fachhochschule, Departement Gesundheit, Zürich, Switzerland

1

Introduction: The demand for health care services is continuously increasing, boosted by demographic, epidemiological, economic and social changes (EVD, 2010). Nursing graduates from vocational and higher education levels are required to meet this challenge, and to provide sustainable care services. In 2014, 1,718 nurses graduated with a diploma in nursing (“Höhere Fachschule”) (Swiss Red Cross, 2015). Part of them are well prepared to advance their scope of practice with a BScN degree at a University of Applied Science (UAS) and foster innovative patient care. Some even pursue their career with an MScN, focusing either on practice development and project management, or on advanced practice roles for particularly complex patient situations, needed namely in geriatric and home care settings. Methods: We aimed to identify the progress in clinical practice from a nursing diploma to a BScN degree. Firstly, we conducted an analysis of the clinical structures of our students. Secondly, we analyzed students’ transfer reports written at the end of the BScN program. The CanMEDS model was used as reference for analysis (Canadian model for medical students’ core competencies). It had also been used as framework for competencies in health care degree programs at Swiss UAS. The seven roles are: experts in care, communicator, team worker, manager, health advocate, scholar, professional (Ledergerber et al., 2009, p. 17). Results: 10 reports from a BScN class were analysed using a coding scheme. Students demonstrate their systematic identification of patients’ needs by applying clinical assessment skills. They are able to evaluate deteriorations more precisely and faster (expert). They communicate appreciatively and professionally (communicator). They extend their view from individuals to families and understand them as co-producers (health advocate). Students control the nursing process in challenging patient situations. They plan evidence-based interventions and reflect patient outcomes (professional). Students are able to support colleagues from other professions, including other students (scholar), and they are accountable within interprofessional teams (team worker). Conclusion: Pursuing a BScN program after an associate degree, and studying within a didactics of research curriculum, enables and empowers graduates to meet new challenges and work in innovative care models. Disclosure: Ursina Baumgartner: Employment or Leadership Position: Führungsposition.

Stein H.1, Bob R.1, Dürkop H.1, Erck C.2, Kämpfe D.3, Kvasnicka H.-M.4, Martens H.5, Roth A.6, Streubel A.6 Pathodiagnostik Berlin, Referenzzentrum Berlin für Hämatopathologie, Berlin, Germany, 2Helmholtz-Zentrum für Infektionsforschung GmbH, Braunschweig, Germany, 3Praxis für Onkologie, Lüdenscheid, Germany, 4Senckenbergisches Institut für Pathologie, Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany, 5Synaptic Systems GmbH, Göttingen, Germany, 6Medizinisches Versorgungszentrum am Helios Klinikum Emil von Behring, Labor für molekulare Diagnostik und Mikrobiologie, Berlin, Germany 1

Introduction: Recent advances in myeloproliferative neoplasms discovered CALRETICULIN (CALR) mutations as a major actor in these disorders. In contrast to JAK2 mutations being mainly associated with Polycythaemia vera, CALR mutations are only associated with primary myelofibrosis (PMF) and essential thrombocythaemia (ET) CALR mutations are present in the majority of primary myelofibrosis and essential thrombocythaemia patients lacking JAK2 and MPL mutations. Since these mutations are absent from reactive bone marrow (BM) lesions their presence indicates ET or PMF. So far these mutations are detectable only by molecular assays. Their molecular detection is cumbersome because of the great CALR-mutation heterogeneity. Therefore, the availability of a simple assay would be of great help. All CALR mutations reported lead to a frameshift generating a new 36 amino acid C-terminus. Methods and results: We generated a monoclonal antibody (CAL2) to the C-neoterminus by immunizing mice with a representative peptide and compared its performance with Sanger sequencing data in 173 MPNs and other BM diseases. There was 100% correlation between the molecular and the immunhistological CALR-mutation-detection. Conclusion: Thus, the immunodetection of CALR-mutations by the CAL2 antibody is a specific, sensitive, rapid, simple and low-cost method. The prevalent immunostaining of megakaryocytes confirms the strongest expression of mutated CALRETICULIN in megakaryocytes. Disclosure: Harald Stein: Employment or Leadership Position: Geschäftsführer der Optistain GmbH; Stock Ownership: Anteilseigner an der Optistain GmbH; Honoraria: Anmeldung eingereicht; Expert Testimony: Eigenfinazierung. V81

Analyses of 357 patients with PMF, pET-MF and pPV-MF – a retrospective field study Junghanß C.1, Springer G.2, Jentsch-Ullrich K.3, Balser C.4, Hauch U.5, Kisro J.K.6, Dengler J.7, Kiewe P.8, Königsmann M.9, Köchling G.10, Ostermann G.11, Kragl B.1 Universitätsmedizin Rostock, Zentrum für Innere Medizin, Klinik III – Hämatologie, Onkologie und Palliativmedizin, Rostock, Germany, 2HämatoOnkologische Schwerpunktpraxis Stuttgart am Hauptbahnhof, Stuttgart, Germany, 3Schwerpunktpraxis für Hämatologie und Onkologie, Magdeburg, Germany, 4Praxis für Innere Medizin, Hämatologie & internistische Onkologie, Marburg, Germany, 5Onkologische Praxis, Erfurt, Germany, 6Ambulantes Tumorzentrum, Neustadt, Germany, 7Schwerpunktpraxis Onkologie, Heilbronn, Germany, 8MVZ Onkologischer Schwerpunkt am Oskar-Helene-Heim, Berlin, Germany, 9Onkologisches Ambulanzzentrum am Diakoniekrankenhaus Henriettenstiftung gGmbH, Hannover, Germany, 10Onkologische Schwerpunktpraxis, Villingen, Germany, 11Novartis Pharma GmbH, Business Unit Oncology, Nürnberg, Germany 1

Introduction: Primary Myelofibrosis (PMF), post essential thrombocythemia (pET) and post polycythemia vera (pPV) MF are rare diseases. Therapeutical options have significantly improved by the development of novel drugs, improvement of allogeneic stem cell transplantation (SCT) and supportive care. Whereas PMF, pET-MF and pPV-MF clinical trial

20

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

participants are well characterized regarding personal, disease and treatment issues this is not the case for the general MF population. Here, we performed a questionnaire poll in multiple institutions gathering data on 357 patients. Methods: A two sided questionnaire asking for general patient and disease specific information as symptoms, splenomegaly, prognostic factors, past and current treatment and blood values, degree of MF in bone marrow and transfusion frequency was developed. Participating centers (n = 15) were mostly private offices in Germany. Data of 357 patients was documented by the participants and analyzed centrally. A median of 20 pts (range 10–60 pts) per center were documented in 2013/14. Results: Gender distribution was: 48% female, 52% male. Age at first diagnosis was: < 50y (10%), 51–60y (21%), 61–70y (33%), >70y (36%). Most pts had PMF (69%), pPV-MF (13%), pET-MF (18%). Major disease durations: 1–5y (42%) and >5y (45%). Thrombocythopenia: <50GPT/l: 6.3%, 50 to <100GPT/l: 14.4% and thrombocytosis ≥450GPT/l: 20.7%. Hemoglobin [g/dl]: ≥10 (60%), 8–10.0 (28%), <8 (12%). 16% of all pts had WBC >25.000/µl and 13% showed circulation blasts in the peripheral blood, respectively. Further DIPPS characteristics were: >65y (72%) and constitutional symptoms 24%. Common symptoms included splenomegaly (66%), decreased fitness (41%) and weight loss (23%). Pruritus was present in 5% and night sweats in 13% of all pts. Concomitant diseases were: cardiac 57%, GI 12%, diabetes 10%, secondary neoplasia 9%. Non-medical treatments were rare: SCT 4.2%, splenectomy 2.8%, spleen irradiation 3.6%. Medical treatment included cytostatics (37%), anticoagulation (30%), JAK-inhibitors (27%), none (20%). Only 36% of all pts received any RBC transfusion, 12% received >50 units. Conclusion: Daily practice MF pts share several characteristics with the COMFORT trial cohort. As expected the diseases were not as progressed as in the trials. Interestingly male gender predominance was not as pronounced in our study. SCT is a rarely used treatment whereas JAK2 inhibitors are frequently used. Disclosure: No conflict of interest disclosed. V82

Interferon gamma secreted by activated NK cells has a negative impact on the treatment with tyrosine kinase inhibitors (TKI) in CML cells Held S.A.E.1, Heine A.1, Schönberg K.1, Riethausen K.1, Daecke S.N.1, Wolf D.1, Brossart P.1 Universitätsklinikum Bonn, Medizinische Klinik III, Bonn, Germany

1

Introduction: Chronic myeloid leukemia (CML) is a myeloproliferative disease which resolves in the constitutive activity of the BCR-ABL protein. Since the introduction of imatinib as first line therapy for CML, the overall survival of patients could be significantly extended. However, mutations of the BCR-ABL protein and environmental effects mediated by adhesion molecules, cytokines and growth factors were shown to induce resistance to TKIs. Therefore, we determined the role of activated NK cells and analyzed the effects of released IFNγ on CML cells. Methods: CML cell lines and primary cells from patients with CML were treated with imatinib or nilotinib and incubated with IL12/IL18 activated or naïve NK cells in transwell experiments. Induction of apoptosis was analyzed by flow cytometry. In addition, IFNγ was used together with TKIs and apoptotic cell death as well as MHC class I expression was determined. Furthermore, effects of IFNα in contrast to IFNγ on apoptosis induction and MHC class I expression were examined. Immunoblotting was performed to analyze the involved pathways. Results: Incubation of CML cells in transwell assays with activated NK cells resulted in significant inhibition of the apoptosis induction by imatinib and nilotinib. To further understand the mechanisms mediating these effects, CML cells were treated with IFNγ in addition to the TKIs. In line with the results from previous experiments, IFNγ reduced the rate of cell death mediated by imatinib and nilotinib. Interestingly, in contrast to IFNα, IFNγ inhibited the TKI-induced downregulation of MHC-class I molecules on CML cells. In line with these results, Western blot analysis

22

Oncol Res Treat 2015;38(suppl 5):1–270

of K-562 and Kyo-1 cells incubated with imatinib or nilotinib in combination with IFNγ showed an increased activation of Jak2 in contrast to TKI treatment alone. To complete this, the activation of Jak2 by IFNγ resulted in an elevated nuclear expression of the transcription factor Runx1 which is supposed to play an important role in CML disease persistence. Conclusion: Our results demonstrate that IFNγ released by activated NK cells can interfere with the action of TKIs in CML cells by reducing the TKI mediated apoptosis induction followed by an increased activation of Jak2 and expression of Runx1. These results might have important implications on the therapy in CML as well as in the development of immunotherapeutic approaches. Disclosure: No conflict of interest disclosed. V83

The observed incidence of cardiovascular ischemic events in dasatinib-treated patients in clinical trials compared with the expected incidence based on general external populations Saglio G.1, le Coutre P.2, Cortes J.3, Mayer J.4, Rowlings P.5, Mahon F.-X.6, Kroog G.7, Gooden K.7, Subar M.7, Shah N.P.8 University of Torino, Ospedale San Luigi Gonzaga, Orbassano-Torino, Italy, Charité, Campus Virchow Klinikum, Universitätsmedizin Berlin, Berlin, Germany, 3The University of Texas MD Anderson Cancer Center, Houston, United States, 4University Hospital Brno, Brno, Czech Republic, 5Calvary Mater Newcastle Hospital, University of Newcastle, Newcastle, Australia, 6Centre Hospitalier Universitaire de Bordeaux and Institut Bergonié, Bordeaux, France, 7 Bristol-Myers Squibb, Princeton, United States, 8UCSF School of Medicine, San Francisco, United States 1 2

Introduction: Arterial ischemic events have been observed with some BCR-ABL1-targeted tyrosine kinase inhibitors (TKIs); therefore, the incidence of cardiovascular (CV) ischemic events in dasatinib-treated patients was assessed. Methods: CV ischemic events were assessed in a pooled population of patients with Ph+ leukemia across 11 clinical trials (N = 2712) and the two Phase 3 studies DASISION (N = 519, newly diagnosed patients with CML) and READY (N = 1518, patients with prostate cancer). Standardized incidence rates (SIRs) were calculated to determine if the number of observed events was different than expected compared with reference populations (general, N = 116,000,000; males, N = 56,000,000; CML patients, N = 16,000; prostate cancer patients, N = 530,000) from Truven’s MarketScan Commercial Claims database, 2008–2013, narrowed to mimic trial eligibility. SIRs were calculated by dividing the observed number of events in dasatinib-treated patients by the expected number of events, based on dasatinib exposure and reference population event rates. Results: Within the pooled population, 96 patients (4%) had CV ischemic events. In DASISION, 10 dasatinib- and 4 imatinib-treated patients had any grade CV ischemic events. In READY, 18 patients in the dasatinib arm and 9 patients in the placebo arm had any grade CV ischemic events. The majority of patients with an event had history and/or risk factors for atherosclerosis (77/96 in the pooled population; 8/10 with dasatinib in DASISION). Most cardiovascular ischemic events occurred within 1 year of dasatinib initiation (69/96 patients in the pooled population and 7/10 patients in DASISION). Based on SIRs, the observed number of cardiovascular ischemic events in dasatinib-treated patients was not higher than expected (95% confidence intervals were less than or included 1). SIRs should be interpreted with caution due to the limitations of the comparison between clinical and claims data (e.g. coding differences and surveillance bias). Conclusions: The incidence of CV ischemic events in dasatinib-treated patients was low in this analysis. The majority of patients with an event had history and/or risk factors with most events occurring early and largely restricted to 1 year after initiating therapy. SIRs suggest the number of CV ischemic events among dasatinib-treated patients was not higher than expected.

Abstracts


CONTENTS AUTHOR INDEX

Disclosure: Giuseppe Saglio: Financing of Scientific Research: Novartis, Bristol-Myers Squibb, ARIAD, Pfizer Neil Shah: Expert Testimony: Bristol-Myers Squibb, ARIAD, Pfizer.

Table 1. Efficacy

V84

CA180–034 final study results: Seven-year (yr) follow-up of chronic-phase chronic myeloid leukemia (CML-CP) patients (pts) with imatinib resistance or -intolerance receiving dasatinib, a 2nd-generation BCR-ABL tyrosine kinase inhibitor (TKI) Müller M.C.1, Shah N.P.2, Rousselot P.3, Schiffer C.4, Rea D.5, Cortes J.6, Mohamed H.7, Healey D.7, Kantarjian H.6, Hochhaus A.8, Saglio G.9 Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany, 2UCSF School of Medicine, San Francisco, United States, 3Hôpital Mignot, University Versailles Saint-Quentin-en-Yvelines, Le Chesnay, France, 4 Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, United States, 5Hôpital Saint-Louis, Paris, France, 6The University of Texas MD Anderson Cancer Center, Houston, United States, 7Bristol-Myers Squibb, Princeton, United States, 8Universitätsklinikum, Jena, Germany, 9University of Torino, Ospedale San Luigi Gonzaga, Orbassano-Torino, Italy 1

Introduction: CA180–034 (NCT00123474), a prospective, randomized phase 3 study, compared the dose and schedule of dasatinib, a BCR-ABL TKI, for optimal benefit/risk ratio among imatinib-resistant or -intolerant CML-CP pts. Previous study results demonstrated significant efficacy of dasatinib, leading to its current approval at 100 mg once daily (QD) in this population. We present here the final 7-yr analysis for efficacy and safety in CA180–034, which is the longest follow-up of any 2nd-generation BCRABL TKI to date. Methods: Pts (n = 670) were randomized to dasatinib: 100 mg QD (n = 167); 50 mg twice daily (BID; n = 168); 140 mg QD (n = 167); or 70 mg BID (n = 168). To manage inadequate response or adverse events (AEs), dose modifications (total daily dose [TDD] of 20–180 mg) were allowed. After 2 yrs, the protocol was amended to allow switching to a QD regimen with the same TDD. Results: Approximately 55% (50 mg BID) and 51% (70 mg BID) of pts treated after the protocol amendment switched to QD dosing by the last recorded dose. Median duration of therapy was longer in the 100 mg QD arm (37.4 months [mos]) vs other dose groups. Rates for major molecular response (MMR), progression-free survival (PFS), and overall survival (OS) were similar across doses (Table). In an exploratory landmark analysis, pts in the 100 mg QD arm with BCR-ABL ≤10% (on the International Scale) at 3 mos had improved 7-yr PFS and OS rates relative to pts with BCR-ABL >10% (PFS [95% CI]: 56% [43–67] vs 21% [10–34]; OS [95% CI]: 72% [60–81] vs 56% [42–68], respectively). For 100 mg QD, most non-hematologic and hematologic AEs (all grades) first occurred within 24 mos. Fewer grade 3–4 AEs occurred at 100 mg QD. With a minimum of 7 yrs of follow-up, pleural effusion rate was lower in the 100 mg QD arm vs other dose groups (any grade [severe]: 28% [5%] vs 35% [8%], respectively). Overall, 128 pts (19%) received ≥7 yrs of dasatinib therapy. Conclusions: Long-term follow-up with dasatinib demonstrates durable efficacy and benefit in imatinib-resistant or -intolerant pts with CML-CP, particularly if achieving BCR-ABL ≤10% at 3 mos. Dasatinib is well-tolerated, with most AEs occurring early in therapy; however, pleural effusion occurred throughout 7 yrs of treatment. No new safety signals were detected.

Abstracts

100 mg QD (n = 167)

50 mg BID (n = 168)

140 mg QD (n = 167)

70 mg BID (n = 168)

MMR in assessed treated pts, n (%)

73 (46)

70 (44)

68 (44)

69 (46)

PFS at 7 yrs,% (95% CI)

42 (33–51)

44 (35–53)

38 (30–47)

44 (35–52)

OS at 7 yrs,% (95% CI)

65 (56–72)

70 (62–77)

73 (65–80)

68 (60–75)

Disclosure: Martin Müller: Expert Testimony: Bristol-Myers Squibb, Novartis, ARIAD; Other Financial Relationships: Personal fees & non-financial support – Bristol-Myers Squibb, Novartis, ARIAD Giuseppe Saglio: Financing of Scientific Research: Novartis, Bristol-Myers Squibb, ARIAD, Pfizer. V85

Long-term 5-year results of the phase 3 DASISION trial (dasatinib versus imatinib in newly diagnosed chronic myeloid leukemia in chronic phase [CML-CP]) Stegelmann F.1, Cortes J.2, Saglio G.3, Baccarani M.4, Kantarjian H.2, Mayer J.5, Boqué C.6, Shah N.P.7, Chuah C.8, Casanova L.9, Narayanan G.10, Bradley-Garelik B.11, Manos G.11, Hochhaus A.12 Universitätsklinikum, Ulm, Germany, 2The University of Texas MD Anderson Cancer Center, Houston, United States, 3University of Torino, Ospedale San Luigi Gonzaga, Orbassano-Torino, Italy, 4S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy, 5University Hospital, Brno, Czech Republic, 6Institut Català d’Oncologia, Hospital Duran i Reynals, L’Hospitalet, Barcelona, Spain, 7 UCSF School of Medicine, San Francisco, United States, 8Singapore General Hospital and Duke-National University of Singapore Graduate Medical School, Singapore, Singapore, 9Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 10Regional Cancer Centre, Medical College, Thiruvananthapuram, Kerala, India, 11Bristol-Myers Squibb, Princeton, United States, 12Universitätsklinikum, Jena, Germany 1

Introduction: The randomized, phase 3 DASISION trial has demonstrated improved efficacy with dasatinib (DAS) versus imatinib (IM) in newly diagnosed CML-CP patients (pts). We report here the results of the final 5-year (yr) analysis of long-term efficacy and safety outcomes. Methods: Treatment-naïve CML-CP pts were randomized to receive DAS 100 mg once daily (QD; n = 259) or IM 400 mg QD (n = 260) and were followed for the predefined minimum of 5 yrs. Results: At the time of study closure, 61% and 63% of DAS- and IM-treated pts, respectively, were still on initial therapy. Confirmed complete cytogenetic response (cCCyR), major molecular response (MMR), and MR4.5 rates by 5 yrs continued to be higher for DAS versus IM (cCCyR: 83% vs 78%; MMR: 76% vs 64%; MR4.5: 42% vs 33%). DAS had fewer cases of transformation to accelerated/blast phase (AP/BP) on study or after discontinuation (4.6%) compared with IM (7.3%). Overall 5-yr progression-free survival (PFS) and overall survival (OS) rates were high and similar for DAS (PFS: 85%; OS: 91%) and IM (PFS: 86%; OS: 90%). A higher proportion of pts on DAS achieved BCR-ABL ≤10% at 3 months (84%) compared with IM (64%). The improved PFS, OS, and lower rates of transformation to AP/BP previously reported for pts who achieved BCRABL ≤10% versus >10% at 3 months were maintained at 5 yrs for both treatment arms. Fifteen pts on DAS and 19 pts on IM had BCR-ABL mutations. No unexpected safety events were identified in either treatment arm at 5 yrs; however, additional DAS-treated pts experienced a first occurrence of pleural effusion (PE). Overall, 74 DAS-treated pts (29%) experienced PE (grade1/2=67; no grade 5). Fifteen pts discontinued DAS due to PE. Arterial ischemic events overall were not common (DAS: n = 12; IM: n = 6). Six of 14 DAS-treated pts with pulmonary hypertension discontinued therapy. No DAS-treated pts had World Health Organization Group

Oncol Res Treat 2015;38(suppl 5):1–270

23


CONTENTS AUTHOR INDEX

1 pulmonary arterial hypertension (confirmed by right heart catheterization) or peripheral arterial occlusive events. Conclusion: At 5 yrs, DAS 100 mg QD demonstrated superior efficacy compared with IM 400 mg QD as first-line therapy for CML. More pts on DAS achieved BCR-ABL ≤10% at 3 months, and there were higher cumulative rates of response and a lower rate of transformation on DAS. PFS and OS were similar for both treatment arms. No new safety signals were reported. These consistent results confirm that DAS offers meaningful advantages over IM for pts with newly diagnosed CML-CP. Disclosure: Frank Stegelmann: Other Financial Relationships: Bristol-Myers Squibb, Novartis, Pfizer, ARIAD Andreas Hochhaus: Expert Testimony: ARIAD, Bristol-Myers Squibb, Merck Sharpe & Dohme, Novartis, Pfizer.

Wissenschaftliches Symposium Long Term Survivorship V86

Social and financial consequences of cancer Seifart U.1 Klinik Sonnenblick, Marburg, Germany

1

The number of the long time “Cancer-Survivor” does increase, because of rising incidence and improved prognosis of patients (pts) [1]. In germany nearly 50% of the 1. 4 million cancer pts are younger than 65y. Only about 50% of them will return to work completely [2]. This means that every year approximately 100,000 tumour pts will face the question whether and how return to work. The unfitness for work results in an increased risk of long-term unemployment or retirement. This may become for the affected persons a considerable financial distress (FD) which is linked with depressed quality of life and social isolation [3]. FD can show a significantly bigger load than possible physical or psychic side effects in particular for young adults, independent of the income. The diminished social standing may also correlates with the prognosis our pts. For germany an evaluation of ten cancer registers showed that tumour pts from income-strong regions showed a better 5 year overall survival rates than cancer pts from socially weaker areas [4]. The risk a longer-term acquisition inability correlates, on one hand with the tumour entity, but also with the intensity of the therapy. A risk decrease can be reached by professional help. This is available in the oncological rehabilitation [5], but also to the cancer advice centres. References: 1 Robert Koch-Institut (Hrsg.). Beiträge zur Gesundheitsberichterstattung des Bundes. Krebs in Deutschland 2009/2010. 9. Ausgabe, 2013. 2 Mehnert A Employment and work-related issues in cancer survivors. Crit Rev Oncol Hematol 2011;77(2):109–130. 3 Shankaran V et al. Risk factors for financial hardship in patients receiving adjuvant chemotherapy for colon cancer: a population-based exploratory analysis. J Clin Oncol 2012;30(14):1608–1614. 4 Jansen, L., et al for the GEKID Cancer Survival Working Group. Socioeconomic deprivation and cancer survival in Germany: An ecological analysis in 200 districts in Germany. Int J Cancer 2014;134: 2951–2960. 5 Mehnert A, et al . Predictors of employment among cancer survivors after medical rehabilitation – a prospective study. Scand J Work Environ Health 2013;39(1):76–87. Disclosure: No conflict of interest disclosed.

24

Oncol Res Treat 2015;38(suppl 5):1–270

V87

Cognitive dysfunction with regard to return to work Rick O.1 Klinik Reinhardshöhe, Onkologie, Bad Wildungen, Germany

1

Cognitive dyfunction (CD) is a short-term, long-term or permanent functional disorder in attention, concentration, thought processes, memory (especially short-term memory), learning ability and the ability to perform complex tasks. Depending on the investigated tumor disease, the prevalence of CD varies significantly between 12% and 68%, it can increase to 80% 6 months after the end of chemotherapy and is described up to 20 years after the cancer treatment. CD is most likely multifactorial. Besides the actual tumor, genetic conditions, age and the anti-tumor therapy (chemotherapy, surgery, radiation therapy) play the key role. The interplay of these components ultimately determines the severity of the cognitive disorder. The correlation between CD and participation in the labor market is only insufficiently investigated and returns partly contradictory outcomes. In patients with Hodgkin lymphoma, CD has a negative impact on the return to working life. Another study has shown that cancer patients with CD resume professional activity less frequently than patients without CD. In a study of 45 patients with breast cancer, the negative influence of CD on return to work could not be determined. Despite the inhomogeneous and little evidence-based data, it can be assumed that especially patients with special professional requirements for cognitive function can be prevented from resuming professional activity by this kind of disorder. Moderate instances of memory impairment can make the performing of simple professional activities impossible. In particular disorders of the ultra-short-term memory (up to about sixty minutes back) normally impact performance to such a significant extent that the pursuit of a professional activity is no longer possible. It is essential to clarify, on a case-by-case basis, which individual professional activities are reduced or prevented by CD and how. This is best done by means of a differentiated vocational true-to-life stress test in the context of oncological rehabilitation. Disclosure: No conflict of interest disclosed. V88

Sport and therapeutic exercise in the long-term treatment of young cancer patients Koenig V.1 Klinik Bad Oexen, Bad Oeynhausen, Germany

1

Functional impairments and therapy-related disorders may develop after cancer therapy, if intensifed therapeutic strategies are used, as is often the case in adolescents and young adults (AYA). Multimodal therapy (combination of surgery, radiotherapy and cytotoxic chemotherapy) might lead to general fatigue. To recover from cancer therapy, medical doctors often recommend physical inactivity. As a consequence, secondary problems may develop. However, for a sustanied physical improvement after cancer treatment, an adequate level of physical training is necessary, thus supporting the adjustment of body structures (i.e. strengthening of muscle fibers, increase in cardiac and pulmonary efficiency etc.). It has been observed that regular exercising after cancer therapy improves overall survival. In AYA physical fitness is markedly reduced after antineoplastic treatment compared to older adults, as AYA receive more aggressive therapeutic regimens. AYA should therefore undergo an intensified physical training program compared to older patients. This can be achieved within an age-specific group rehabilitation in a specialist clinic (in-patient program). A high-level exercise schedule within a peer group is effective by the competition initiated among the group members. Typically, such a program takes 4 weeks and is only a first step on the way back to life. For a complete recovery up to the old performance levels, the physical training program has to be continued for at least several months. The patients

Abstracts


CONTENTS AUTHOR INDEX

should be informed and motivated accordingly. Similar programs can also be applied in a single exercise environment in an out-patient setting, with reduced efficiency (more motivation needed to be successful). Disclosure: No conflict of interest disclosed. V89

Impact and effects of sleep disturbances in younger cancer patients Strik H.1 Universität Marburg, Neurologische Klinik, Marburg, Germany

1

Sleep disturbances, daytime sleepiness and psychiatric symptoms are fequent, but only rarely asked for in daily oncological routine. Most probably, patients and their treating physicians consider sleep disturbances to be inevitable in cancer disease. Usually, oncologists ask their patients for tumor specific symptoms like pain, dyspnoea, loss of appetite or nausea. The quality of sleep is often disregarded. Disturbances of sleep can have various reasons. Undesired effects of tumor therapy can inhibit the sleep as well as supportive treatment. Most frequently, sleep disturbances are caused by psychosocial distress which can be depression as well as financial worries. Moreover, indirect effects of the tumor disease or its treatment on the hormonal regulation regulation of sleep are possible. The extensive treatment of tumor diseases should therefore include a systematic assessment of sleep disturbances and treatment of their causes. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Kopf-Hals-Tumore I V90

Prospective multicenter phase II study of the anti-EGFR (epidermal growth factor receptor) antibody panitumumab (P) in patients with platinum pre-treated, advanced head and neck squamous cell cancer (HNSCC) Siano M.1,2, Molinari F.3, Martin V.3, Mach N.4, Früh M.1, Crippa S.3, Ghielmini M.5, Frattini M.3, Espeli V.5

and 39.4% (13 pts) stable diseases (SD), resulting in a clinical benefit rate of 45.5%. Both PRs occurred in platinum-resistant pts. Median PFS was 3.0 months (95% CI: 1.8, 5.5) and median duration of clinical benefit (PR+SD) 8.4 months (95% CI: 3.6, 17.7). Pts with PR were free of progression for 8 and 14 months. 54.5% of pts had only grade 1–2 toxicity, while 27.3% experienced grade 3–4 P-related adverse events. Cutaneous rash was the most frequent (27/33 pts in total [81.8%], of Grade 3 in 2 [6.1%], requiring treatment withdrawal in 1pt), followed by hypomagnesaemia (22/33 pts [66.7%], of Grade 3–4 in 5 [15.2%]). One pt died after the first P administration, presumably due to drug-related severe alveolitis since no other cause was identified, and one pt experienced serious hypokalaemia grade 4. P was in general well tolerated and no infusion-related reactions occurred. Conclusion: We present the first efficacy and safety results of P monotherapy in platinum pre-treated HNSCC pts. P was well tolerated showing antitumor activity and remarkable duration of clinical benefit. Toxicity profile and convenience support consideration of P in this setting. Disclosure: No conflict of interest disclosed. V91

Subgroup analysis according to differentiated thyroid cancer histology in phase 3 (SELECT) trial of lenvatinib Elisei R.1, Schlumberger M.2, Tahara M.3, Robinson B.4, Brose M.5, Dutcus C.6, Zhu J.6, Newbold K.7, Kiyota N.8, Kim S.-B.9, Sherman S.10, Wirth L.11 Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy, 2Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and University Paris-Sud, Villejuif, France, 3Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan, 4Kolling Institute of Medical Research, University of Sydney, New South Wales, Australia, 5Department of Otorhinolaryngology: Head and Neck Surgery, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, United States, 6Eisai Inc, Woodcliff Lake, United States, 7The Royal Marsden National Health Service Trust, London, United Kingdom, 8Department of Medical Oncology and Hematology. Kobe University Hospital, Kobe, Japan, 9 Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Republic of, 10Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, United States, 11Department of Medicine, Massachusetts General Hospital, Boston, United States 1

Introduction: We performed a multicenter phase II study with the fully human anti-EGFR-antibody P, administered as a single agent in platinum-pretreated HNSCC patients (pts). We present the first safety and efficacy data in this pt population. Material and methods: Recurrent or metastatic HNSCC pts previously treated with platinum containing chemotherapy in four tertiary Swiss cancer centers were included. Previous anti-EGFR-antibody treatment (EAB) was allowed if pts had no progression during or within six months after therapy. P was administered intravenously every two weeks at a dose of 6mg/kg. Primary endpoint was response rate (RR), secondary endpoints were duration of response (DR), progression-free survival (PFS), and safety. Adverse events (AEs) were graded following the NCI Common Terminology Criteria (CTCAE V3.0). Tumor response was assessed every 8 weeks according to RECIST V1.1. Tumor tissue samples were collected and centrally analyzed for potential, predictive biomarkers. Results: 33 pts received in total 151 and a median of 4 (range1–16) cycles of P. Ten pts (30.3%) were EAB pre-treated and 26 pts (78.8%) were platinum resistant or refractory. RR was 6.1% with two partial responses (PR)

Background: The previously reported results of the SELECT trial demonstrated that lenvatinib significantly prolonged progression-free survival (PFS) in patients with 131I-refractory differentiated thyroid cancer (RRDTC) compared with placebo. Median PFS in lenvatinib and placebo were 18.3 and 3.6 months, respectively (hazard ratio [HR]: 0.21, 99% confidence interval [CI]: 0.14–0.31; P < 0.0001). This subgroup analysis of SELECT trial examined clinical outcomes of each histological characteristics. Methods: In the SELECT trial, 392 patients with RR-DTC were enrolled and randomized 2:1 to lenvatinib (24 mg/day, 28-day cycle) or placebo. The primary endpoint was PFS and the secondary endpoints were including objective response rate (ORR), overall survival (OS) and safety. This analysis examines the clinical outcomes of prespecified subgroups divided based on histology: papillary thyroid cancer (PTC, n = 259) and follicular thyroid cancer (FTC, n = 133). Results: PTC patients treated with lenvatinib and placebo had median PFS of 16.4 months and 3.5 months, respectively (HR: 0.27; 95% CI: 0.19– 0.38). FTC patients treated with lenvatinib had a median PFS that had not yet been reached vs 3.7 months for placebo (HR: 0.10; 95% CI: 0.05–0.19). ORR results of the patients treated with lenvatinib were 63.9% (PTC) and 66.3% (FTC). Although the OS advantage with lenvatinib vs placebo had detected with no significant difference (HR: 0.73; 95% CI: 0.50–1.07) in the overall population, possibly due to the cross-over design, the clinically meaningful OS advantage was observed in lenvatinib-treated FTC group (HR: 0.41; CI: 0.18–0.97). A treatment-related TEAE (TR-TEAE) and grade ≥3 TR-TEAEs occurred in 96.4% and 76.3% in PTC patients and 98.9% and 75.0% in FTC patients, respectively.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Cantonal Hospital St. Gallen, Clinic of Oncology / Hematology, St. Gallen, Switzerland, 2Fondazione IRCCS Istituto Nazionale dei Tumori, Head and Neck Medical Oncology Unit, Milan, Italy, 3Institute of Pathology, Laboratory of Molecular Diagnostics, Locarno, Switzerland, 4Geneva University Hospital, Clinical Research Unit of the Dr. Henri Dubois-Ferrière Dinu Lipatti Foundation Oncology Center, Geneva, Switzerland, 5Oncology Institute of Southern Switzerland, Medical Oncology, Bellinzona, Switzerland 1

25


CONTENTS AUTHOR INDEX

Conclusions: Clinical benefits of lenvatinib treatment were observed in both examined PTC and FTC subgroups. These results such as the promising response in the FTC patients warrant further investigation to enrich the evidence. Disclosure: Rosella Elisei: Advisory Role: Bayer, AstraZeneca, Genzyme; Financing of Scientific Research: Bayer, AstraZeneca, Genzyme; Other Financial Relationships: Fees from: Genzyme, Bayer, Sobi/Exelixis, AstraZeneca Lori Wirth: Other Financial Relationships: Fees from: Eisai, Novartis.

V93

Prognostic and predictive factors correlated with treatment outcomes for radioactive Iodine-refractory differentiated thyroid cancer (RAI-rDTC) patients receiving sorafenib or placebo on the phase III DECISION trial Paschke R.1, Schlumberger M.2, Elisei R.3, Pacini F.4, Jarzab B.5, Giannetta L.6, Bastholt L.7, de la Fouchardiere C.8, Worden F.P.9, Shong Y.K.10, Smit J.W.11, Kappeler C.12, Molnar I.13, Brose M.S.14 Leipzig University, Department for Endocrinology and Nephrology, Leipzig, Germany, 2Gustave Roussy, Villejuif, France, 3University of Pisa, Department of Clinical and Experimental Medicine, Pisa, Italy, 4University of Siena, Unit of Endocrinology, Siena, Italy, 5Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Gliwice, Poland, 6Ospedale Niguarda Ca’ Granda, Milan, Italy, 7Odense University Hospital, Odense, Denmark, 8Hospices CivilsCentre Anticancéreux, Consortium Cancer Thyroïdien, Lyon, France, 9University of Michigan Health System, Ann Arbor, United States, 10Asan Medicine Center, Seoul, Korea, Republic of, 11Radboud University Nijmegen Medical Center, Department of Internal Medicine, Nijmegen, Netherlands, 12Bayer Pharma AG, Berlin, Germany, 13Bayer HealthCare Pharmaceuticals, Whippany, United States, 14 Abramson Cancer Center of the University of Pennsylvania, Department of Otorhinolaryngology, Head and Neck Surgery, Philadelphia, United States 1

V92

Phase II explorative trial to prospectively investigate predictive molecular biomarkers for efficacy of panitumumab (P) in platinum-pretreated head and neck squamous cell cancer (HNSCC) Siano M.1,2, Molinari F.3, Martin V.3, Mach N.4, Früh M.1, Crippa S.3, Ghielmini M.5, Espeli V.5, Frattini M.3 Cantonal Hospital St. Gallen, Clinic of Oncology / Hematology, St. Gallen, Switzerland, 2Fondazione IRCCS Istituto Nazionale dei Tumori, Head and Neck Medical Oncology Unit, Milan, Italy, 3Institute of Pathology, Laboratory of Molecular Diagnostics, Locarno, Switzerland, 4Geneva University Hospital, Clinical Research Unit of the Dr. Henri Dubois-Ferrière Dinu Lipatti Foundation Oncology Center, Geneva, Switzerland, 5Oncology Institute of Southern Switzerland, Medical Oncology, Bellinzona, Switzerland 1

Introduction: Monoclonal antibodies directed against EGFR (E-Mab) were extensively investigated in HNSCC. Even if clinical efficacy in terms of survival was shown to be marginal, there seems to be a subgroup of pts with clear benefit and durable responses. So far, no single biomarkers or biomarker combination pattern was found to identify this subgroup. The aim of this study was to analyze pre-specified biomarkers in a prospective phase II trial with P in HNSCC pts. Methods: Pts with platinum-pretreated HNSCC included in a prospective phase II multicenter trial exploring P as 2nd line treatment consented for molecular biomarker analysis on available tumor tissue. As P is not active through indirect mechanisms like ADCC (antibody dependent cellular cytotoxicity), it seems to be an ideal candidate to examine the pure effect on EGFR pathway, held responsible for durable responses. To find a predictive marker pattern for response, a central laboratory investigated the following markers: KRAS, NRAS, HRAS, PI3KCA, BRAF gene mutations by Sanger sequencing; EGFR gene status by FISH; HPV genotyping. Results: 25 pts consented for the biomarker sub-study. Tumor tissue was available in all pts. One case was excluded for bad quality of DNA. Two uncommon KRAS mutations (G48E, T50I, 8%) and 3 (12.5%) PIK3CA mutations (all E545K) were detected; all the other genes were wild type. HPV high-risk 16 was found in 9 (37.5%) and EGFR copy number gain (CNG) in 12 pts (50%). No correlation between response and molecular alterations was observed, with the exception of EGFR: all 3 PR patients showed EGFR CNG, a feature not identified in SD or PD patients. Conclusion: EGFR CNG by FISH may identify HNSCC patients who will benefit from P administration and may be a useful marker to predict efficacy of P. This preliminary observation needs to be confirmed in a larger series. The cascade of MAP kinases (RAS, BRAF) was not predictive. The role of PIK3CA mutations remains to be elucidated. Disclosure: Marco Siano: Other Financial Relationships: Amgen stellte das Medikament zur Verfügung für die Studie Milo Frattini: Other Financial Relationships: Amgen stellte das Medikament zur Verfügung für die Studie.

26

Oncol Res Treat 2015;38(suppl 5):1–270

Background: The DECISION trial established that sorafenib prolonged progression-free survival (PFS) compared to placebo in patients with progressive RAI-rDTC (Lancet 2014). Here we sought to identify prognostic and predictive factors correlated with treatment outcomes. Methods: Multivariate Cox proportional hazards models adjusted for treatment effect and subgroup analyses defined by maximum target lesion size and existence of disease-related symptoms were used to explore the relationship between clinical baseline variables and PFS. Patients were deemed symptomatic at entry if they had symptoms/findings consistent with thyroid cancer reported in the medical history or pre-treatment adverse event dataset. Results: A total of 417 patients were randomized to receive placebo (n = 210) or sorafenib (n = 207). Multivariate Cox model analyses indicated that lower maximum individual target lesion size, lower number of lesions, thyroglobulin levels at baseline less than the median (486 ng/ ml) and region Asia versus Europe and North America were prognostic for longer PFS in placebo patients and in all patients when adjusted for treatment. Subgroup analyses indicated that patients whose maximum individual target tumor size were <1.5 cm had longer PFS and appeared to have less benefit from sorafenib treatment than patients with lesions ≥1.5 cm. Lesions ≥1.5 cm as well as lung metastases were predictive for better treatment effect with sorafenib. Both symptomatic and asymptomatic patients at entry had improved PFS following treatment with sorafenib. Conclusions: Maximum tumor size, number of lesions, thyroglobulin levels at baseline and geographic regions were prognostic for longer PFS in RAI-rDTC patients. Individual tumor size ≥1.5 cm and lung-only metastases were predictive for better treatment effect with sorafenib. Patients appeared to benefit from sorafenib treatment irrespective of disease-related symptoms at baseline. Thus, based on these post hoc exploratory analyses, patients with progressive RAI-rDTC and maximum tumor size <1.5 cm appear to have a good prognosis and may be candidates for “watch and wait” before initiating sorafenib. Disclosure: Ralf Paschke: Advisory Role: Bayer Pharma AG, Eisai; Financing of Scientific Research: Bayer Pharma AG, Eisai Marcia Brose: Advisory Role: Bayer HealthCare Pharmaceuticals, Exelixis, Onyx Pharmaceuticals; Financing of Scientific Research: Bayer HealthCare Pharmaceuticals; Expert Testimony: Bayer HealthCare Pharmaceuticals, Exelixis, Eisai, Novartis, Roche/Genentech.

Abstracts


CONTENTS AUTHOR INDEX

V94

Characterization of tumor-associated B cell subsets in head and neck squamous cell carcinoma Lechner A.1,2, Rothschild S.I.2,3, Schlößer H.A.2,4, Thelen M.2, Shimabukuro-Vornhagen A.2, Beutner D.1, von Bergwelt-Baildon M.S.2 Uniklinik Köln, Klinik und Poliklinik für Hals-, Nasen- und Ohrenheilkunde, Köln, Germany, 2Uniklinik Köln, Klinik I für Innere Medizin, Cologne Interventional Immunology, Köln, Germany, 3Universitätsspital Basel, Medizinische Onkologie, Basel, Switzerland, 4Uniklinik Köln, Klinik und Poliklinik für Allgemein-, Viszeralund Tumorchirurgie, Köln, Germany 1

Background: Recent evidence suggests that B cells can both promote and inhibit the development and progression of tumors. Tumor-infiltrating B cells have been described in a large variety of solid tumors including head and neck squamous cell carcinoma (HNSCC). However, there has not been a detailed phenotypic characterization of the different B cell subsets in HNSCC. The aim of this study was to characterize the composition of the B cell infiltrates in HNSCC in order to gain further insight into their role. Methods: In this study we characterized B cell subsets in primary tumors (n = 30), blood (n = 30) and non-cancerous mucosa (n = 10) of 30 previously untreated patients diagnosed with HNSCC. Furthermore, peripheral blood from samples of age-matched healthy donors (n = 10) was included as control. B cell subsets were analyzed by 10-color flow cytometry. Results: Tumor-infiltrating B cells (CD19+) can be found in the majority of HNSCC samples. HNSCC tissue contained a significantly higher percentage of activated B cells (CD19+/CD20+/CD86+) compared to healthy controls, non-cancerous mucosa of HNSCC patients and peripheral blood of HNSCC patients. The percentage of memory B cells (CD19+/CD20+/ IgD-/CD27+) was significantly higher in tumor tissue than in the peripheral blood of cancer patients. Moreover, we found a higher rate of regulatory B cells (CD24high/CD38high or TIM-1+) in tumor tissue compared to peripheral blood of healthy controls and tumor patients as well as non-cancerous mucosa. Conclusion: B cells constitute a significant proportion of the immune infiltrate in HNSCC. The B cell infiltrate of HNSCC is characterized by an accumulation of CD86+ activated B cells and memory B cells which is suggestive of an antigen recognizing phenotype and a specific effector B-cell response. However, tumors are also infiltrated by a considerable number of regulatory B cells. Disclosure: No conflict of interest disclosed.

[or other sensitive imaging]), size of the M-spike, IgA isotype, abnormal serum free light chain (sFLC) ratio, degree of PCosis, circulation and aberrancy of BMPCs and immunoparesis. Ultra-HR-SMM is now redefined as MM with BM PCosis >60%, sFLC ratio >100 and >1 focal lesion on wbMRI. The number of these redefined pts is small (10–15%), but important to unravel, because their risk of progression is substantial (≥80% within 2 years). Pts with HR FISH cytogenetics (CG) (del17p, t(4;14), gain 1q21) are not yet considered for early treatment, because groups are heterogeneous and other RF than CG deemed to weight higher. Albeit pts with ultra HR-SMM are now considered as MM and treated as such (Table 1), concerns do exist that earlier therapy may increase the risk of selecting resistant clones, induce side effects and costs. Therefore, the even more accurate identification of pts, who most likely benefit from interventions, needs to be performed, and clinical judgement and careful discussion of pros and cons of treatment initiation undertaken. For the greater majority of SMM pts, the standard of care remains observation until development of symptomatic MM occurs. The latter group remains to be observed every 3–6 months and is encouraged to participate in clinical trials. Tab. 1.

Disclosure: No conflict of interest disclosed. V97

Treatment should immediately be started in every patients with high risk smoldering myeloma (SMM): No – defending the NO position Ludwig H.1

Wissenschaftliches Symposium

Hoch-Risiko Smouldering Myelom (SMM): Pro und Kontra unverzügliche Behandlung V96

Treatment should immediately be started in every patient with high risk smouldering myeloma (SMM) Engelhardt M.1, Wäsch R.1 University of Freiburg Medical Center, Department of Hematology, Oncology & Stem Cell Transplantation, Freiburg, Germany 1

SMM is an asymptomatic clonal plasma cell (PC) disorder and bridges MGUS and MM, based on higher levels of circulating monoclonal immunoglobulin (IG) and bone marrow (BM) plasmocytosis (PCosis) without end-organ damage. Until the PETHEMA-GEM group reported fewer MM-related events and better overall survival (OS) among patients (pts) with high-risk (HR)-SMM treated with lenalidomide-dexamethasone, prior studies failed to show improved OS, assessing bisphosphonates, melphalan, prednisone or thalidomide as compared to observation. Risk factors (RF) of HR-SMM have now been defined (Table 1), and a subset of ultra-HR pts are reclassified by the International Myeloma Working Group as MM, based on biomarkers that identify pts with a >80% risk of progression. Commonly defined RF for SMM progression are the number of bone lesions, e.g. abnormal whole-body (wb) magnetic resonance imaging (MRI

28

Oncol Res Treat 2015;38(suppl 5):1–270

Wilhelminenspital, Wilhelminen Cancer Research Institute, Vienna, Austria

1

Introduction: Based on improved PFS and OS obtained in one small study (Metaeos MV et al., NEJM 2013) with immediate start of Lenalidomide/Dexamethasone therapy as compared to starting treatment only at time of clinical evident progression, the IMWG recommended immediate therapy in all patients with newly defined high risk SMM ( ≥ 1 focal lesions in MRI, or FLC ratio >100 and involved FL >100mg/dl). Methods: Review of available evidence (Medline, ASH, EHA, IMWG, ASCO meetings) and risk calculation Findings: Hillengass L. et al. showed that ≥1 focal lesion of >5 mm size detected in MRI associate with a 70% rate of progression, a finding which was confirmed by another small study (Kastridis E. et al.) showing similar results (69% rate of progression). The FLC ratio risk model (FLC ratio >100 and involved FLC concentration of >100 mg/dl) was associated with an 82% risk of progression after 2 years (Larsen JT et al., 2013), but when the risk for progression was assessed in another patient cohort using a FLC ratio >100 alone, only 53% of patients with SMM had progressed after 2 years (Waxmann AJ et al., 2015). Risk modelling: These data show a 2 year rate of progression between 70 and 82% using either one of the risk models. Assuming a linear rate of transformation into active myeloma, the progression rate will be 20%, 40%, 60, and 80% after 6, 12, 18, and 24 months, and the rate of unnecessary treatment will amount to 80%, 60%, 40%, and 20% at the given time points, indicating significant overtreatment. Calculating the area under

Abstracts


CONTENTS AUTHOR INDEX

the curve patients require treatment and above the curve when patients do not require therapy shows a ratio of 40% to 60%, meaning that 60% of the time therapy was administered without medical need. Conclusion: The recommendation to start therapy in every patient with high risk SMM, given the toxicity and burden of therapy, does not accord with one of the most important principles in medicine, namely ‘primum non nocere’. Modelling shows that roughly 60% of the treatment will be administered to patients without clinical signs of active myeloma if the recommendations are followed. This reality must be presented to and discussed with the individual patient to enable him to make an informed decision. In case the patient is unable or unwilling to decide treatment should be withheld, but the patient needs to adhere to careful regular control examinations in short intervals by an experienced myeloma specialist. Disclosure: No conflict of interest disclosed.

Fortbildung

Sarkom – Vergleich der Behandlungskonzepte V104

Osteosarcoma: A step backwards for poor responders? What´s next? Bielack S.1, Cooperative Osteosarkom-Studiengruppe COSS Klinikum Stuttgart – Olgahospital, Päd. Onkologie, Hämatologie, Immunologie, Stuttgart, Germany 1

For the past 35 years, the treatment of osteosarcoma has been based upon surgery of the primary tumor and, if present, primary metastases plus pre- and postoperative chemotherapy. Many international groups would consider a combination of high-dose methotrexate, adriamycin (doxorubicin), and cisplatin (MAP) as standard. It has long been proven that the extent of histologic response of the primary tumor to upfront preoperative chemotherapy is correlated with survival expectancies, and patients whose tumors still contain more than 10% viable cells after standard induction carry an unfavorable prognosis. Until recently, no prospective randomized trial evaluating whether postoperative treatment modifications might improve the prognosis of these poor responders had been performed. The European and American Osteosarcoma Study EURAMOS1 (NCT00134030) filled this void. Poor responders to 10 weeks of MAP induction were randomized to either continue with MAP postoperatively until protocol week 29 or to receive postoperative MAP augmented by high-dose ifosfamide and etoposide until protocol week 40 (MAPIE). A total of 2.260 patients from 17 countries were registered into EURAMOS1, 1.059 of these were considered poor responders, of whom 618 participated in the poor response randomization (Marina et al., CTOS 2014, paper 032). At the predefined analysis of the primary endpoint, event-free survival (EFS), higher acute toxicities were observed with the augmented, experimental MAIE chemotherapy, as was an increased incidence of secondary acute leukemias. There was no evidence that the more toxic MAPIE was more efficacious than MAP, and EFS rates did not differ between arms. The four multinational osteosarcoma groups collaborating in EURAMOS1 therefore recommend to not use MAPIE in case of poor response to MAP, but to continue with MAP regardless of histologic response – the standard treatment which will still result in cure in well over 40% even of poor responders. Meanwhile, the search for novel treatment options continues. As of yet, no substance which might be available for large-scale, multi-institutional, multi-national phase III osteosarcoma trials has been identified. Current international efforts aim to describe genomic and immunologic targets for novel treatment approaches and to translate these findings from bench to bedside. The EURAMOS consortium intends to initiate their next large joint front-line trial once an agent suitable for such an approach is identified

V105

Position papers and treatment recommendations for chordomas and desmoid tumours Kasper B.1 Mannheim University Medical Center, Interdisciplinary Tumor Center, Sarcoma Unit, Mannheim, Germany 1

Desmoid-type fibromatosis (DF) is a rare disease characterized by a monoclonal, fibroblastic proliferation and a variable and often unpredictable clinical course. The incidence is less than 3% of all soft tissue sarcomas. Due to the rarity and the long natural history of the disease, the level of evidence as it is available for more common tumour types is currently beyond reach for DF. Especially in the context of systemic treatment options, only few phase II trials are available, and most published data arises from retrospective case reports or small series. There is no published phase III randomized clinical study on DF and no established or evidence-based treatment approach is available as of today. With this background information in mind, a European joint effort was initiated bringing together sarcoma experts from the European Organisation for Research and Treatment of Cancer (EORTC) / Soft Tissue and Bone Sarcoma Group (STBSG) with patients and patient advocates from Sarcoma Patients EuroNet (SPAEN), and has led to the development of consensus recommendations based on patients’ and professionals’ expertise – for the first time ever in this disease. As a surrogate, a position paper has been published (Kasper B et al.: Eur J Cancer 2015;51:127–136). Chordomas are very rare malignant bone tumours that have had a shortage of effective treatments for a long time. Although new treatment options are available for both local and metastatic disease, the degree of uncertainty in selecting the most appropriate treatment remains high and their adoption in daily life remains inconsistent. As a blueprint of the above described procedure, a consensus meeting on chordoma management was hosted by the European Society for Medical Oncology (ESMO) within their regular meeting updating the clinical practice guidelines on sarcomas. This meeting included more than 40 chordoma experts from several disciplines with the contribution and sponsorship of the Chordoma Foundation, a global patient advocacy group. The consensus reached at that meeting has recently been published in a position paper (Stacchiotti S et al.: Lancet Oncol 2015;16:e71–83). The bullet points of the position papers and this innovative process of developing treatment recommendations and a therapeutic algorithm in rare diseases will be presented. Disclosure: No conflict of interest disclosed.

Fortbildung

Hodentumore – bestes Management V106

Management seminoma Stage IIB: Radiation therapy Papachristofilou A.1 Universitätsspital Basel, Radioonkologie, Basel, Switzerland

1

Optimal management of seminoma stage IIB remains a controversial issue. Standard radiation therapy of the paraaortal and ipsilateral pelvic lymphatic pathways leads to excellent local control, at the cost of distant recurrence and potential long term sequlae of large volume irradiation. In the past years innovative techniques have lowered the risk of excessive radiation dose to the kidneys and bowel, yet the problem of disease recurrence beyond the irradiated volume remains a challenge. Innovative combined treatment concepts encompassing reduced volume as well as possibly reduced dose radiation therapy and less intensive systemic treatment may provide the optimal future treatment option.

Disclosure: Stefan Bielack: Advisory Role: Bayer, Celgene, IDM/Takeda Millennium, Lilly, Merck & Co., Roche.

Disclosure: No conflict of interest disclosed.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

29


CONTENTS AUTHOR INDEX

V107

Management of seminoma stage IIB: Chemotherapy is the treatment of choice Honecker F.1 Tumor- und Brustzentrum ZeTuP, St. Gallen, Switzerland

1

Seminoma stage II B is defined by a N2 status diagnosed by abdominal CT scan. Definition of N2 is either a retroperitoneal lymph node mass of more than 2 but less than 5 cm, or multiple lymph nodes, any one mass extending between 2 and 5 cm in greatest dimension. The optimal management of stage IIB is controversial, and data from randomized trials is lacking. Treatment recommendations are as follows: the European Association of Urology (EAU) currently considers chemotherapy (CT) with 3 × PEB or 4 × PE as an alternative to radiotherapy (RT) with 36 Gy (in 2 Gy fractions) to paraaortic and ipsilateral iliac lymph nodes [1], whereas the European Society of Medical Oncology (ESMO) clinical practice guidelines from 2013 consider CT as the optimal choice [2]. Notably, when CT is considered, it must be performed according to the current standard, as use of 4 cycles of single agent carboplatin was insufficient and resulted in higher relapse rates [3]. Retroperitoneal lymph node dissection is highly experimental, and should not be carried out outside clinical trials. A systematic review and meta-analysis evaluated data from studies using either RT or CT in stage IIA/B [4]. 4 prospective and 9 retrospective studies, none of them randomized, and with a total of 890 patients (607 RT, 283 CT) were identified. Pooled relapse rate (RR) was 0.11 for RT (CI 0.08–0.14) and 0.08 for CT (CI 0.01–0.15). For assessment of overall mortality, 8 studies were available, and outcome was excellent with pooled mortality rates of 0.02 for RT (CI < 0.01–0.04) and 0.01 for CT (CI < 0.01–0.02). Despite statistical equivalence between RT and CT, a trend towards lower relapse rates and lower incidences of late toxicities, especially less GI toxicity and secondary cancers was observed. This is “suggesting an erosion of equipoise in favor of CT for stage IIB seminoma” [4]. An ongoing trial is assessing the efficacy and safety of combination therapy with single-agent carboplatin plus involved field RT in this situation. The trial is open in multiple centers in Germany and Switzerland, and referral of all suitable patients with stage II seminoma is strongly recommended. References: 1 Albers P et al.: Uroweb 2015, http://uroweb.org. 2 Oldenburg J et al.: Ann Oncol 2013;24(Suppl 6):vi125-vi132. 3 Krege S et al.: Ann Oncol 2006;17:276–280. 4 Giannatempo P et al.: Ann Oncol 2015;26:657–668. Disclosure: No conflict of interest disclosed. V108

Management Seminoma IIB – pro retroperitoneal surgery Albers P.1 Universitätsklinikum Düsseldorf, Klinik für Urologie, Düsseldorf, Germany

1

Es gibt zur retroperitonealen Resektion der Metastasen im klinischen Stadium II der Seminome als singuläre Therapieoption kaum zitierbare Daten. In einer Serie aus Magdeburg, in der über 63 Patienten mit retroperitonealer Lymphadenektomie (RLA) bei Seminomen berichtet wird, fällt eine “in-field“ Rezidivrate von 9.5% (6/63) und eine Gesamtrezidivrate für Seminome ab Stadium IIC von 58% (7/12) auf. Dabei wurden 23 der 63 Patienten nach der RLA adjuvant therapiert. Die Operation wurde daher ab 1985 nur noch zurückhaltend durchgeführt. In der aktuellen Literatur findet man entsprechend nur noch Fallberichte (n = 4, Hu, 2015; n = 4, Mezvrishvili, 2006). Pathophysiologisch gibt es keine Gründe, Seminome bezüglich operativer Strategien vor Chemotherapie grundsätzlich anders zu behandeln als Nicht-Seminome. Auch Seminome folgen in ihrer Metastasierung sehr lange den Lymphabflußwegen und metastasieren erst spät hämatogen. Aus klinischer Sicht gibt es aber folgende Gründe, metastasierte Seminome im Stadium IIA und IIB nach Ablatio testis primär retroperitoneal zu operieren:

30

Oncol Res Treat 2015;38(suppl 5):1–270

1. marker-negative Tumoren mit unklarer Histologie, die einer Punktion nicht zugänglich sind 2. imperative Indikationen bzw. Kontraindikationen zur Chemotherapie wie z. B. Niereninsuffizienz (autosomal dominante Zystennieren) 3. Spät-Toxizität der Chemotherapie 4. Spät-Toxizität der Radiotherapie Es müssen grundsätzlich drei Szenarien unterschieden werden: a) primär diagnostizierte Tumoren im Stadium II, b) Rezidiv unter Überwachung c) Rezidiv nach 1 × Carboplatin adjuvant. Die Standardtherapie dieser Stadien wäre 3 × BEP Chemotherapie oder Radiatio mit 36 Gy. Beide Therapieoptionen haben eine messbare Langzeittoxizität und werden undifferenziert und unabhängig von Größe, Lokalisation sowie Markerhöhe eingesetzt. Besonders Rezidive unter Überwachung und nach Carboplatin werden aber üblicherweise sehr früh in der Nachsorge erkannt (Tumordurchmesser oft <1.5 cm), so dass (insbesondere bei marker-negativen Rezidiven) im Rahmen prospektiver Studien eine primäre Resektion dieser kleinvolumigen Metastasen als singuläre Therapieoption zur Vermeidung von Spät-Toxizitäten geprüft werden sollte. Im Rezidivfall bleiben die bisherigen Primärtherapien als Rezidivtherapien erhalten. Disclosure: No conflict of interest disclosed. V110

Palliative treatment of testicular germ cell cancer Oechsle K.1 Universitätsklinikum Hamburg-Eppendorf, Onkologisches Zentrum, II. Medizinische Klinik und Poliklinik, Hamburg, Germany 1

Introduction: Although 70–80% of germ cell cancer patients can be cured even in metastatic disease, prognosis remains poor in patients with multiple relapses during or after cisplatin-based chemotherapy or relapse after high-dose chemotherapy. Methods: A systematic overview on current literature on treatment of patients with refractory or multiply relapsed germ cell cancer including recommendations for daily clinical practice will be presented. Results: Single agent activity with response rates of about 20% could be observed for orally applied etopside, paclitaxel, gemcitabine and, oxaliplatin (Einhorn 1991, Motzer 1994, Bokemeyer 1999, Kollmannsberger 2002). Until today, the evaluation of targeted therapy only revealed some marginal activity for sunitinib with remission rates of 10% (Feldman 2010, Oechsle/Honecker 2011) and thalidomide with marker, but nor morphologic remissions in about 30% (Rick 2006). None of these agents showed significant impact on overall survival when applied as monotherapy. Various studies on combination chemotherapy regimen combining two of the three agents, paclitaxel, gemcitabine, and oxaliplatin, achieved response rates of about 20–40% with overall survival times of 6–8 months (e.g. Hinton 2001, De Giorgi 2006, Einhorn 2007, Kollmannsberger 2004). In the following, a further German study evaluated the triple combination chemotherapy with gemcitabine, oxaliplatin plus paclitaxel (GOP-regimen) and demonstrated the highest response rate for the present of 51% in these heavily pretreated patients: 78% after high-dose chemotherapy (Bokemeyer 2008). After additional secondary surgery, complete remission was achieved in 20% of patients. Further follow-up confirmed long term survival >2 years in 17% of these patients (Oechsle 2011). Similar effects were present in a retrospective study on the triple combination chemotherapy of cisplatin, gemcitabine, and paclitaxel resulting in a 2 year overall survival rate of 30% after complete secondary surgery. (Necchi 2014) Conclusions: Results of single agent treatments are still disappointing and only of palliative nature in patients with multiply relapsed or refractory germ cell cancer. The evaluation of new substances remains if enormous relevance for these patients. Triple combination with the GOP regimen could result in long term survival in about 15–20% of these patients if complete secondary surgery of all residual lesion could be achieved. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag MDS klinisch I V114

Prognostic significance of rare single abnormalities in Myelodysplastic Syndromes Schanz J.1, Tüchler H.2, Solé F.3, Sanz G.4, Garcia-Manero G.5, LeBeau M.6, Bennett J.M.7, Slovak M.L.8, Fenaux P.9, Malcovati L.10, Cazzola M.10, Pfeilstöcker M.11, Valent P.12, Ohyashiki K.13, Levis A.14, Sekeres M.15, Tauro S.16, Magalhaes S.17, Van de Loosdrecht A.18, Cermak J.19, Lübbert M.20, Stauder R.21, Germing U.22, Greenberg P.23, Haase D.24, On behalf of the International Work Group for Prognosis in MDS (IWG-PM) Universitätsmedizin Göttingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany, 2Hanusch Hospital, Boltzmann Institute for Leukemia Research, Vienna, Austria, 3Institut de Recerca contra la Leucèmia Josep Carreras, Barcelona, Spain, 4Hospital Universitario La Fe, Valencia, Spain, 5 The University of Texas, MD Anderson Cancer Center, Houston, United States, 6 University of Chicago Comprehensive Cancer Research Center, Chicago, United States, 7James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, United States, 8Quest Diagnostics Nichols Institute, Chantilly, United States, 9Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris (AP-HP)/University Paris XIII, Bobigny, France, 10Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy, 11Hanusch Hospital and L. Boltzmann Cluster Oncology, Vienna, Austria, 12Medical University of Vienna, Vienna, Austria, 13 First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan, 14Fondazione Italiana Sindromi Mielodisplastiche c/o SS Antonio e Biagio Hospital, Allessandria, Italy, 15Cleveland Clinic Taussig Cancer Institute, Cleveland, United States, 16University of Dundee, Dundee, United Kingdom, 17 Federal University of Ceara, Fortaleza, Brazil, 18Department of Hematology, VU University Medical Center, Amsterdam, Netherlands, 19Institute of Hematology and Blood Transfusion, Prague, Czech Republic, 20University of Freiburg Medical Center, Freiburg, Germany, 21University of Innsbruck, Innsbruck, Austria, 22Heinrich-Heine University Hospital, Düsseldorf, Germany, 23Division of Hematology, Stanford University Cancer Center, Stanford, United States, 24 Universitätsmedizin, Göttingen, Germany 1

Introduction: The IPSS (Greenberg et al., 1997) and its recently published revision, the IPSS-R (Greenberg et al., 2012), defines the standard in risk stratification of patients with Myelodysplastic Syndromes (MDS). However, due to its low frequency, the prognostic impact of rare abnormalities remains unclear as yet. These abnormalities are coalesced in one group in the IPSS and IPSS-R. The main goal of the study presented here was to analyse the type, frequency and prognosis of rare single abnormalities in a large cohort of patients with primary, untreated MDS. Methods: To date, we identified 382 pts. with rare single abnormalities derived from the international database of the IWG-PM, containing more than 7000 patients. A specific group was defined as having at least n = 5 cases with the same abnormality. The participating centers (in numerically order) were: Spain (n = 109; 28.5%), MD Anderson Cancer Center (73; 19.1%), IMRAW (43; 11.3%), France (34; 8.9%), Pavia (25; 6.5%), Vienna (21; 5.5%), Duesseldorf (16; 4.2%), Japan (14; 3.7%), Italy (11; 2.9%), Cleveland (10; 2.6%), Dundee (9; 2.4%), Brazil (8; 2.1%), Netherlands (5; 1.3%), Czech (2, 0.5%), Freiburg (1; 0.3%) and Innsbruck (1; 0.3%). For all abnormalities detected, the median overall- (OS) and AML-free survival (AFS) was calculated. Results: Rare single abnormalities detected were: +1/+1q/dup1q (n = 9; 2.4%), t(1;var)(12; 3.1%), del(1q)(5; 1.3%), der(1;7)(19; 5.0%), del(3p)(7; 1.8%), t(3;var)(5; 1.3%), t(5q)(8; 2.1%), del(6q)(7; 1.8%); -9/del(9q)(15; 3.9%), t(11q23)(6; 1.6%), +13 (6; 1.6%), -13/del(13q)(20; 5.2%), +14(5; 1.3%), del(16q)(7; 1.8%), del(17p)(7; 1.8%), -19 (5; 1.3%), +21 (16; 4.2), -21/del(21q)(6; 1.6%), -X (7; 1.8%), and +mar (12; 3.1%). The remaining 198 patients (51.8%) showed rare abnormalities occurring in less than 5 patients. The median overall survival as well as the AML-free survival in each category will be presented in detail. Furthermore, a proposal to integrate these abnormalities in the cytogenetic module of the IPSS-R will be presented.

Abstracts

Conclusions: Due to their low frequency, rare abnormalities in MDS can only detected based on large, international cooperation projects. In the following study, the identification and prognostic impact of rare abnormalities, uninfluenced by therapy or additional abnormalities was comprehensively analyzed. The results will lead to a further refinement of the cytogenetic prognostic classification in patients with MDS. Disclosure: No conflict of interest disclosed. V115

CMML and treatment with azacitidine in a routine care setting in Germany: Data from the second planned interim analysis of the non-interventional study “observational study on treatment patterns with VIDAZA®” Illmer T.1, Prange-Krex G.2, Lück A.3, Lollert A.4, Schwarzer A.5, Bruch H.-R.6, Bachinger A.7, Weiligmann C.8, Steudel C.8, Platzbecker U.9 Onkologische Gemeinschaftspraxis, Dresden, Germany, 2Gemeinschaftspraxis Mohm und Prange-Krex, Dresden, Germany, 3Zentrum für Onkologie und Urologie in Rostock, Rostock, Germany, 4Gemeinschaftspraxis Dr. Neise & Dr. Lollert, Krefeld, Germany, 5Gemeinschaftspraxis Hämatologie/Onkologie, Leipzig, Germany, 6Praxis Bonn – Schwerpunktpraxis, Bonn, Germany, 7 ClinAssess GmbH, Leverkusen, Germany, 8Celgene GmbH, München, Germany, 9 Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany 1

Introduction: Approval of azacitidine (AZA) in chronic myelomonocytic leukemia (CMML) is based on the pivotal AZA001 trial. This prospectively, randomized trial, which has shown prolonged overall survival (OS) in MDS patients (pts), included few CMML pts (n = 11). Additionally, the approval in CMML is restricted to the non-myeloproliferative disease. Here we present data from CMML pts out of the second interim analysis of a non-interventional multicentre trial in Germany. The aim of the study was to prospectively collect data on the clinical usage of AZA in the real world setting. Methods: At the time of data cutoff (04/16/2015) 440 pts of 660 planned pts were enrolled in 66 German sites and observed for a maximum of 12 months with an optional one year of follow-up. A comprehensive set of data on demography, disease characteristics, AZA treatment regimen including dosage and duration, reasons for discontinuation and other factors were collected and analyzed. Survival data were estimated according to Kaplan Meier methodology. Results: At the time of this analysis, 44 pts diagnosed with CMML had been recruited and were evaluable. The majority were elderly pts (median age of 75.5 years [range 57–89]) and treated with the approved schedule for 7 days (75.0%). Five pts (11.4%) received AZA in second or further line of treatment. Overall response rate (CR, CR-BM, PR or HI) was 36.4% (n = 16) and an additional 25.0% (n = 11) achieved SD while in 38.6% (n = 17) no response assessment data were available at the time of data cutoff. Six pts (13.6%) achieved a complete or partial cytogenetic response. One year survival rate was 59.1% (95% CI; 43.2–73.7). Treatment beyond one year was documented in one quarter of pts (n = 11). Main reasons for treatment discontinuation were death (27.3%, n = 9), pt request (15.2%, n = 5) and multiple reasons (24.2%, n = 8). Half of all pts experienced serious adverse events (54.5%, n = 24). The most common serious adverse events during treatment were general disorders and administration site conditions (18.2%, n = 8). Conclusion: This report about 44 CMML pts treated with AZA in a routine care setting in Germany supports evidence that therapy with AZA is effective and safe in CMML. Disclosure: Thomas Illmer: No conflict of interest disclosed. Uwe Platzbecker: Financing of Scientific Research: Celgene GmbH.

Oncol Res Treat 2015;38(suppl 5):1–270

31


CONTENTS AUTHOR INDEX

V116

Progression of chromosomal aberrations correlates with an inferior prognosis in MDS patients with regard to survival and AML progression rates Neukirchen J.1, Nolting A.-C.1, Hildebrandt B.2, Kobbe G.1, Haas R.1, Germing U.1 Uniklinik Düsseldorf, Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University, Düsseldorf, Germany, 2Uniklinik Düsseldorf, Department of Human Genetics, Heinrich-Heine-University, Düsseldorf, Germany 1

Introduction: For MDS patients, several chromosomal abnormalities have been shown to have a significant influence of prognosis. The IPSS as well as in the recently introduced IPSS-R, uses the karyotypes as important factors for evaluation of prognosis. We analyzed the outcome of MDS undergoing subsequent karyotyping during the course of the disease and correlated the results with survival and AML development. Methods: We screened the Duesseldorf MDS registry for those patients that revealed at least two chromosomal analyses during the course of the disease. We then categorized three groups: a) no change of karyotype b) worsening of karyotype, and c) normalization of karyotype and correlated the results with regard to overall survival as well of AML development. Results: We identified in total 595 patients with at least two chromosomal analyses during the course of the disease. The median age was 63 years (17–86 years). 6.2% were categorized as IPSS-R very low, 28.8% as low, 25.5% as intermediate, 19.3% as high and 20.2% as very high. The median number of available karyotypes for each patient was 3 (range 2–25). Looking at all patients with at least two karyotype analyses available, 102 (17.1%) of the patients showed no change of the karyotypes, 292 (49.1%) progressed to a prognostic inferior karyotype and 201 (33.8%) showed a more favorable karyotype during the course of the disease as a result of treatment response. Looking at the AML evolution, time to AML progression was significantly shorter in patients that showed a progression of the karyotype (40 months vs. not reached, p < 0.0001). Accordingly, the survival was with 33 vs. 46 months shorter in the group of patients with karyotype progression (p = 0.03). After exclusion patients that received allogeneic stem cell transplant, the results were comparable. Conclusion: Based on a large database of patients who were karyotyped as least two times, we could demonstrate that in MDS patients clonal evolution is associated with a worse prognosis with regard to survival and AML progression and that the normalization of cytogenetic alterations as a sign of treatment response correlates with a longer survival. These results underline the importance of karyotype analysis for a better prognostication of MDS patients, especially with regard to treatment decision. Disclosure: Judith Neukirchen: No conflict of interest disclosed. Ulrich Germing: Expert Testimony: Celgene, Norvartis, Amgen, Jansen Cilag. V117

Evolution of chromosome 7 material loss in myeloid malignancies Shirneshan K.1, Braulke F.1, Schanz J.1, Haase D.1 Universitätsmedizin Göttingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany 1

Introduction: Loss of chromosome 7 (chr. 7) material (as mon(7) or del(7q)) is one of the most frequent chromosomal abnormalities in patients with acute myelogenous leukemia (AML) and myelodysplastic syndrome (MDS).It has already been demonstrated that the prognostic impact of isolated 7q deletion has to be regarded as prognostically more favorable compared to isolated mon(7) (Corduba et al., 2012, Schanz et al., 2012) and not as unfavorable as described in the IPSS. Method: In 2% of MDS patients we observed a coexistence of mon(7) and del(7q)by FISH analysis of CD34+ peripheral blood cells. Two patients showed an increase of mon (7) clone size parallel to a decline in 7q- clone size during MDS progression.

32

Oncol Res Treat 2015;38(suppl 5):1–270

Results: Here we report three additional MDS cases showing several clones with different chr.7 aberrations. The first case (A.M.): A 72-year old man with suspected MDS. Analysis of bone marrow (BM) revealed MDS RA. Banding analysis of BM cells showed three clones with different chr. 7 abnormalities: del(7q) in 22%, r(7) in 22% and mon(7) in 11% of the metaphases. Second case (A.L.): A 62 year old woman with suspected hematological malignancy. Karyotyping of BM cells revealed: A del(7q) in 12% and r(7) in 15% of metaphases. Third Case (K.B.): A 64 years old MDS Patient with the following karyotype abnormalities: del(7q) in 3%, mon(7) plus only a “naked” centromere of chr. 7 in 47% and complete mon(7) in 20% of BM cells. Conclusion: These results could be a hint that chr. 7 can be prone to karyotype evolution during MDS progression. Due to the existence of the third clone with r(7) and a clone with remaining centromere 7 we assume that r(7) is a transitional stage of karyotype evolution between deletion of 7q and complete monosomy 7 which might result from loss of telomeres in 7q- cells promoting loss of further chromosomal material ending up in a r(7). It is known that ring chromosomes themselves are prone to be lost during cell division. And so finally the entire process ends with complete mon(7). By specific telomere FISH analysis we could confirm the deletion of telomeres in r(7). Our hypothesis could also explain why del(7q) has a better prognosis than mon(7): Del(7q) is occurring earlier during the course of the disease since it is the first step in mon(7)-evolution. To prove this hypothesis, more data from patients with karyotype evolution involving chr. 7 will be accumulated and further specific examinations will be performed. Disclosure: No conflict of interest disclosed. V118

Decitabine given at the 3-day schedule in older patients with refractory anemia with excess blasts in transformation (RAEBt), or low blast count acute myeloid leukemia according to WHO criteria Becker H.1, Suciu S.2, Rüter B.H.1, Platzbecker U.3, Giagounidis A.4, Selleslag D.5, Labar B.6, Germing U.7, Salih H.R.8, Muus P.9, Pflüger K.-H.10, Hagemeijer A.11, Schaefer H.-E.1, Baron F.12, Ganser A.13, Aul C.4, de Witte T.9, Wijermans P.14, Lübbert M.1, EORTC Leukemia Cooperative Group, German MDS Study Group Medical Center – University of Freiburg, Freiburg, Germany, 2EORTC Headquarters, Brussels, Belgium, 3University of Dresden Carl Gustav Carus, Dresden, Germany, 4Marienhospital Düsseldorf, Düsseldorf, Germany, 5AZ St-Jan, Brugge, Belgium, 6University Hospital Center Rebro, Zagreb, Croatia, 7 University Hospital, Düsseldorf, Germany, 8Eberhard Karls University, Tübingen, Germany, 9Radboud University Medical Center, Nijmegen, Netherlands, 10DIAKO, Bremen, Germany, 11University of Leuven, Leuven, Belgium, 12C.H.U. SartTilman, Liège, Belgium, 13Hannover Medical School, Hannover, Germany, 14Haga Ziekenhuis, The Hague, Netherlands 1

Treatment of older patients with high-risk myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) is challenging. In the randomized phase III study 06011 of the EORTC Leukemia Group and German MDS Study Group, we compared decitabine (15 mg/m2 every 8 hours for 3 days, every 6 weeks) with best supportive care (BSC) in patients ≥60 years with MDS by FAB criteria and not eligible for intensive therapy (Lübbert et al.: J Clin Oncol 2011;29:1987–96). In the present analysis, we investigate trial 06011 for the efficacy of decitabine in refractory anemia with excess blasts in transformation (RAEBt), and, after regrouping the study cohort according to WHO criteria, in AML (WHO) with low blast counts. In RAEBt, rates of best response in the decitabine arm (n = 40) were as follows: complete (CR) or partial remission (PR), 15%; hematologic improvement (HI), 15%; progressive disease (PD), 30%. In the BSC arm (n = 35), no patient achieved CR, PR or HI; 80% had PD. The favorable responses in the decitabine arm translated into a significantly longer progression-free survival (PFS; HR 0.30, 95%-CI 0.18–0.51; P < 0.001) and, by trend, longer overall survival (OS; HR 0.68, 95%-CI 0.42–1.11, P = 0.12) than in the BSC arm. Post-progression treatment, particular-

Abstracts


CONTENTS AUTHOR INDEX

ly allogeneic hematopoietic stem cell transplantation (alloHSCT), may influence survival, and the number of patients receiving alloHSCT was imbalanced between the treatment groups. Thus, we censored OS at the time of alloHSCT and observed that the difference between the treatment groups increased (HR 0.63, 95%-CI 0.38–1.04; P = 0.067). This was particularly true for patients aged 60–74 years (HR 0.48, 95%-CI 0.26–0.89; P = 0.018), i.e. potential candidates for alloHSCT after reduced-intensity conditioning. The impact of decitabine on the outcome in RAEBt patients differed from that in patients with refractory anemia with excess blasts (RAEB). After regrouping the study cohort according to the nowadays more widely used WHO classification, AML (WHO) patients in the decitabine arm (n = 27) also had a longer PFS (P = 0.008), and their median OS was 9.8 months compared to 5.9 months in the BSC arm (n = 23). In conclusion, 3-day decitabine is active in MDS and/or AML with 5–30% blood or 20–30% marrow blasts and may be used as bridge to alloHSCT. The 3-day schedule is not feasible in the outpatient setting, but our data point to it as a valid treatment option for hospitalized patients with above features. Disclosure: No conflict of interest disclosed. V119

Underestimation of 12p-deletion in myelodysplastic syndromes? Results from a German diagnostic study in comparison with an international control group Braulke F.1, Müller-Thomas C.2, Götze K.2, Platzbecker U.3, Germing U.4, Hofmann W.-K.5, Giagounides A.A.N.6, Lübbert M.7, Greenberg P.L.8, Bennett J.M.9, Sole F.10, Slovak M.L.11, Ohyashiki K.12, Le Beau M.M.13, Tüchler H.14, Pfeilstöcker M.15, Hildebrandt B.16, Aul C.17, Stauder R.18, Valent P.19, Fonatsch C.20, Bacher U.1, Trümper L.1, Haase D.1, Schanz J.1 Universitätsmedizin Göttingen, Hämatologie und medizinische Onkologie, Göttingen, Germany, 2Technische Universität München, Hämatologie u. Onkologie, München, Germany, 3Uniklinik Dresden, Hämatologie u. Onkologie, Dresden, Germany, 4Universität, Düsseldorf, Germany, 5 Universitätskrankenhaus, Mannheim, Germany, 6Marienhospital, Düsseldorf, Germany, 7Universität Freiburg – Klinik für Innere Medizin I, Freiburg, Germany, 8 Stanford University Cancer Center, Stanford, United States, 9University of Rochester Medical Center, Rochester, United States, 10Institut de Recerca Contra la Leukemia Josep Carreras, Badalona, Spain, 11Sonora Quest Laboratories, Phoenix, United States, 12Tokyo Medical University, Tokyo, Japan, 13University of Chicago, Chicago, United States, 14Hanusch Hospital, Boltzmann Institute for Leukemia Research, Vienna, Austria, 15Hanusch Hospital and L. Boltzmann Cluster Oncology, Vienna, Austria, 16Universität Düsseldorf, Humangenetik, Düsseldorf, Germany, 17St. Johannes Hospital, Duisburg, Germany, 18Innsbruck Medical University, Innsbruck, Austria, 19Division of Hematology and Hemostaseology and Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria, 20Medical University of Vienna, Vienna, Austria 1

Introduction: In myelodysplastic syndromes (MDS), 12p-deletion (del(12p)) as a sole anomaly is a characteristic, but rare abnormality (0.6– 5% at initial diagnosis). It is described to occur as an additional aberration in up to 4% in the context of clonal evolution during the course of the disease. According to current scoring systems, del(12p) is associated with a good to intermediate prognosis as a single aberration. Del(12p) is usually a small interstitial deletion on the short arm of chromosome 12 with a common deleted region between 12p12.2–12p13.1 and ETV6/TEL as the affected gene. Methods: We present the results of a systematic del(12p) testing in our German prospective diagnostic study (clinicaltrials.gov: NCT01355913) on 351 MDS patients in whom CD34+ peripheral blood (PB) cells were analysed by sequential Fluorescence-in-situ-Hybridization (FISH) analyses using a large probe panel including TEL/AML1 (Abbott®, Germany). We compared the CD34+PB-FISH results with the results of chromosome banding analyses (CBA) of bone marrow (BM) metaphases of 2902 previously published MDS patients. Results: Out of 351 MDS patients analysed by CD34+PB-FISH, 28 (7.6%) showed a del(12p) by CD34+PB FISH: 23 at initial screening at the time of study entry, 5 additional patients showed del(12p) later during the course of the study in the context of a karyotype evolution. Most of the del(12p)

Abstracts

patients had primary MDS (85.7%), 4 patients (14.3%) were diagnosed as t-MDS. The clone sizes measured by CD34+PB-FISH and CBA differed significantly for both, non-complex and complex karyotypes involving del(12p), the median clone size of a del(12p) was 65% by CD34+PB-FISH and 100% by CBA of BM metaphases. In contrast, only 45 MDS patients (1.6%) of the CBA control group showed a 12p-deletion. Del(12p) is often associated with other aberrations (93% by CD34+ PB-FISH vs 60% by CBA). Conclusions: The probability to detect del(12p) was significantly higher by FISH as compared to CBA (7.6% by CD34+ PB-FISH vs 1.6% by CBA, p < 0.001). The detection rate of del(12p) could be increased by repeated FISH analyses in a patient over time. It is important for MDS patients to distinguish between a normal karyotype, a single anomaly and double or complex aberrations for the prediction of prognosis according to international prognostic scoring systems. Therefore, we recommend including a probe for del(12p) to common FISH probe panels in MDS. Disclosure: Friederike Braulke: Expert Testimony: Die CD34+FISH-Studie wurde von Celgene und Novartis unterstützt.; Immaterial Conflict of Interests: nein. Julie Schanz: Employment or Leadership Position: nein.; Advisory Role: nein.; Stock Ownership: nein.; Honoraria: nein.; Financing of Scientific Research: nein.; Expert Testimony: Die CD34+FISH-Studie wurde von Celgene und Novartis unterstützt.; Other Financial Relationships: nein.; Immaterial Conflict of Interests: nein.

Fortbildung

Palliativmedizin Entscheidungsprozesse und -kriterien V121

What can the nurse contribute to the decision making process? Monteverde S.1,2 Berner Fachhochschule, Fachbereich Gesundheit, Bern, Switzerland, Universität Zürich, Institut für Biomedizinische Ethik und Medizingeschichte, Zürich, Switzerland 1 2

As in other domains of medical and nursing practice, also in oncology the goal of collaborative decision making is the patient’s health and wellbeing. This is achieved by means of the integration of patient preferences, available scientific evidence and a sensitivity for the relationship between burden and benefit. Patients are encouraged and requested to exercise their right to self-determination and give their consent to the oncological treatment that best fits their values as well as the medical condition. Nevertheless, evidence shows that the threat of the disease and the uncertainties and risks inherent to the medical treatment render the patient and the family particularly vulnerable. Patients and families have multiple needs of information, counseling, assistance and encouragement in making meaningful treatment decisions. These needs are best met when each of the professions involved in the delivery of oncological care plays an active role within the process of shared decision making. Therefore, interprofessional collaboration between doctors and nurses not only covers medical treatment, monitoring and reporting, but also decision making. Due to their continuous presence and proximity to the patients and families, nurses play a crucial role in helping patients to make meaningful decisions. This task is germane to the sphere of co-responsibility shared by doctors and of nurses (see for example Swiss Health Insurance Law (KVG, KLV Art. 7). However, communication research within oncology suggests that the perception of patients, doctors and nurses with regard to the quality of communicative interaction, the understanding of the information delivered, the empathic response and the weighting of risks and benefits of a proposed treatment differs significantly. Similarly, doctors and nurses have different perceptions of the degree to which the other profession is involved in decision making processes. Of particular importance are those decisions that involve the redirection of care towards palliation and quality of life when suffering increases and no benefit is seen for the patient in

Oncol Res Treat 2015;38(suppl 5):1–270

33


CONTENTS AUTHOR INDEX

continuing a curative approach. In redirecting care towards palliation and advance care planning, careful communication, clarification of patient values and readiness for interprofessional negotiation are indispensable. Here, clinical ethics tools like case deliberations, ethics consultations or family conferences are important means to identify meaningful goals of care. Disclosure: No conflict of interest disclosed.

Greiner J.1,2, Schmitt A.3, Hofmann S.1, Götz M.1, Michels B.3, Hückelhoven A.3, Maccari B.4, Lindner D.3, Wang L.3, Wuchter P.3, Mertens T.1, Döhner H.1, Ho A.3, Bunjes D.1, Dreger P.3, Kuball J.5, Schrezenmeier H.4, Schauwecker P.4, Wiesneth M.4, Schmitt M.3 University of Ulm, Department of Internal Medicine III, Ulm, Germany, Diakonie Hospital Stuttgart, Department of Internal Medicine, Stuttgart, Germany, 3University of Heidelberg, Department of Internal Medicine V, Heidelberg, Germany, 4German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Institute of Clinical Transfusion Medicine and Immunogenetics, Ulm, Germany, 5University Medical Center Utrecht, Department of Hematology, Utrecht, Netherlands 1

Freier Vortrag

2

Allogene Transplantation klinisch III V124

The combined impact of amino acid polymorphism at HLADPB1 on T cell alloreactivity defines a functional distance between patient and donor independently predictive of survival after 10/10 matched unrelated HSCT for AML, ALL and MDS Crivello P.1, Beelen D.W.2, Heinold A.3, Heinemann F.M.3, Rebmann V.3, Lindemann M.3, Ottinger H.2, Horn P.A.3, Fleischhauer K.1 Institut für Zelltherapeutische Forschung, Universitätsklinikum, Essen, Germany, 2Klinik für Knochenmarktransplantation, Universitätsklinikum, Essen, Germany, 3Institut für Transfusionsmedizin, Universitätsklinikum, Essen, Germany 1

Introduction: We have previously experimentally determined the differential impact of individual amino acid polymorphisms on the alloreactive T cell response to HLA-DPB1*09:01, translated these findings into numeric “functional distance” (FD) scores for all HLA-DPB1 alleles, and showed that these FD scores correlate with T cell epitope groups determining non-permissive mismatches in unrelated HSCT (Fleischhauer et al.: Lancet Oncol 2012; Crivello et al.: Biol Blood Marrow Transplant 2014). Methods: A cohort of 379 consecutive patients (pts) (age median 56 yrs [18–73 yrs]), who underwent high-resolution HLA-A,B,C,DRB1,DQB1 matched, but DPB1 mismatched unrelated HSCT for AML (n = 272 [72%]), ALL (n = 58 [15%]), or MDS (n = 49 [13%]) at the University Hospital of Essen were included in the analysis. FD scores of DPB1 alleles were calculated as previously described, and the ΔFD scores for each transplant were calculated as the absolute number of [FDpatient-FDdonor]. Receiver Operator Curves (ROC) were constructed to calculate the best cut-off values for the endpoint of overall survival (OS). Results: With a median follow-up of 4 yrs for surviving pts, the 5-yrs OS for pts in early disease stages (n = 158 [42%]) was 56% (95%-confidence interval [CI] 48–64%). For pts in advanced disease stages (n = 221 [58%]) this estimate was 38% (95%-CI 31–45%) (p < 0.0005). ΔFD score distribution ranged from 0.01 to 7.46 with a median of 2.12. ROC analysis indicated stratification into 2 subgroups with ΔFD scores < = 2.665 (n = 252 [66%]) and >2.665 (n = 127 [44%]) as the best predictor. In these subgroups, the 5-yrs OS were 51% (95%-CI 44–57%) and 37% (95%-CI 28–46%), respectively (p < 0.008). In multivariate analysis, independent predictors of OS were time-dependent aGvHD (p < 0.0002) and cGvHD (p < 0.0001), the use of anti-thymocyte globulin (p < 0.0001), patient age (p < 0.004), and the stratified ΔFD score (p < 0.03). Discussion: In this monocentric cohort, the stratified DPB1 ΔFD score was a significant independent risk factor for OS after 10/10 matched unrelated HSCT, possibly owing to the strength of the alloimmune response to mismatched HLA-DPB1 in this setting, which was also reflected by an independently increased non-relapse mortality in pts with ΔFD scores >2.665. These findings could pave the way for the identification of non-permissive DPB1 mismatches in unrelated HSCT based on an experimentally defined numerical weighting system for amino acid sequence polymorphism. Disclosure: No conflict of interest disclosed.

34

V125

Clinical phase I peptide vaccination trial with a CMVpp65derived peptide can prevent and clear CMVpp65 antigenemia after allogeneic hematopoietic stem cell transplantation

Oncol Res Treat 2015;38(suppl 5):1–270

We performed a phase I clinical peptide vaccination trial using an immunodominant viral epitope peptide derived from the phosphoprotein 65 of cytomegalovirus (CMVpp65) for patients after allogeneic stem cell transplantation with a high risk of CMV reactivation, disease and mortality. The CMV phosphoprotein 65 derived nonamer peptide NLVATVPMV has been well characterized as immunogenic. Therefore, we designed a vaccine with 300 microgram of the peptide in an oil-in-water emulsion (Montanide™). Ten CMV-seropositive patients after allogeneic stem cell transplantation received 4 vaccines s.c. at a biweekly interval. Patients were monitored for clinical course as well as CMVpp65 antigenemia. CD8+, Tregs and gamma-delta T cells were analyzed by multi-color flow cytometry. ELISpot assays for Interferon and granzyme B and tetramer assays were performed and correlated to clinical parameters. Peptide vaccination was well tolerated and no drug-related serious adverse events were detected. Eight of nine patients with CMVpp65 antigenemia cleared the CMV after four vaccinations and are still free from viremia until now. One patient received prophylactic vaccination and did not develop viremia. One patient with CMV reactivation showed no clinical response and resented persisting CMV antigenemia. An increase in frequency of CMV specific T-cells was detected in eight patients and frequency of regulatory T-cells was found in a part of the patients. Moreover, the frequency of gamma-delta T cells increased in several but not in all patients with immune and clinical response after vaccination. Taken together, administration of CMVpp65 peptide vaccination for patients after allogeneic stem cell transplantation at high risk for CMV reactivation was save, well tolerated and clinically encouraging. Disclosure: No conflict of interest disclosed. V126

Prognostic impact of aberrant MN1 expression in patients with Acute Myeloid Leukemia (AML) undergoing allogeneic stem cell transplantation in complete remission after NonMyeloablative Conditioning (NMA-SCT) Jentzsch M.1, Bill M.1, Leiblein S.1, Schubert K.1, Wildenberger K.1, Bergmann U.1, Pleß M.1, Weidner H.1, Knyrim M.1, Grimm J.1, Lange T.1,2, Cross M.1, Franke G.-N.1, Pönisch W.1, Vucinic V.1, Behre G.1, Niederwieser D.1, Schwind S.1 Universitätsklinik Leipzig, Hämatologie und internistische Onkologie, Leipzig, Germany, 2Asklepios Klinik Weißenfels, Hämatologie und intern. Onkologie, Weißenfels, Germany 1

Introduction: Meningeoma-1 (MN1) is highly expressed in primitive hematopoietic cells & has dismal prognostic impact in AML patients (pts) treated with chemotherapy. The prognostic significance in AML pts receiving NMA-SCT with a therapeutic effect relying on immunologic graft-versus-leukemia (GvL) effects remains unknown.

Abstracts


CONTENTS AUTHOR INDEX

Patients and methods: We analysed 98 pts (median age 64.1 years [y], range 38.2–75.3y) who received NMA-SCT (90 mg/m2 Fludarabine & 2Gy total body irradiation) in complete remission with pre-treatment bone marrow (BM) material available. Donors were human leukocyte (HLA) matched related (18%) or HLA-matched (57%) or mismatched (24%) unrelated. Medical Research Council (MRC) classification was: 4% favourable, 71% intermediate & 24% adverse. Common surface marker expression was assessed by flow cytometry. Mutation status of NPM1, CEBPA & FLT3-ITD & BAALC & ERG expression were evaluated. MN1 expression was normalized to ABL & the 25th percentile of the normalized gene expression was used to define high & low expressers. Median follow up was 4.3y. Results: High pre-treatment MN1 expression associated with higher CD34+ (P < 0.001), higher CD34+/CD38- (P < 0.001) & by trend higher CD117+ (P = 0.08) BM cell burden, lower white blood count (P = 0.01), lower platelets (P = 0.02), abnormal karyotype (P = 0.003), NPM1 wild type (P < 0.001), high BAALC (P < 0.001) & high ERG expression (P < 0.001). High MN1 expressers had higher cumulative incidence of relapse (CIR) by trend in the entire group (P = 0.07, Figure 1A) & a significantly higher CIR in the MRC intermediate risk subgroup (P = 0.04, Figure 1C). However, this did not translate into shorter overall survival (OS) for the whole or the MRC intermediate risk cohort (P = 0.29 & P = 0.45 respectively, Fig. 1. B&D).

Fig. 1. MN1.

Conclusion: AML pts with high MN1 expression had a higher burden of immature surface marker i.e. CD34 & CD117 & more CD34+/CD38- cells, which harbour the leukemic stem cell population. High MN1 expressers had higher CIR after NMA-SCT, but in contrast to pts after chemotherapy no shorter OS, suggesting that the dismal impact of high MN1 may in part be diminished by the GvL effect after NMA-SCT. Disclosure: No conflict of interest disclosed.

V127

T-cell-replete HLA-haploidentical transplantation using post-transplantation high-dose cyclophosphamide posttransplantation in high-risk and advanced ALL: Feasibilty and early outcome Zoellner A.-K.1, Fritsch S.1, Prevalsek D.1, Köhnke T.1, Hubmann M.1, Lippl S.1, Kruger S.1, Hellmuth J.1, Mumm F.1, Barlow S.1, Ledderose G.1, Subklewe M.1, Spiekermann K.1, Hiddemann W.1, Albert M.2, Schmid C.3, Hausmann A.1,4, Tischer J.1 Ludwig-Maximilians-University Hospital of Munich-Grosshadern, Department III of Internal Medicine, Hematopoietic Stem Cell Transplantation, Munich, Germany, 2Ludwig-Maximilians-University of Munich, Dr. von Haunersches Kinderspital, Munich, Germany, 3Department of Internal Medicine II, Klinikum Augsburg, Augsburg, Germany, 4Department I of Internal Medicine, Klinikum München-Schwabing, Munich, Germany 1

Haematopoietic stem cell transplantation (allo-HSCT) is a potential curative treatment option for patients suffering from high-risk ALL, but less true for patients with advanced disease. However, not in all of our patients a suitable HLA-matched donor could be identified in time. To evaluate the outcome of T-cell-replete (TCR) HLA-haploidentical haematopoietic stem cell transplantation (haplo-HSCT) utilizing high-dose cyclophosphamide post-transplantation in the context of intensification of conditioning in patients with high-risk, relapsed and refractory ALL, we retrospectively analysed the course of 25 patients (B-ALL n = 23, T-ALL n = 2) transplanted between 2010 and 2015 in four German transplant centres. Disease was advanced in 19 patients, including 11 patients with relapse after a first allo-HSCT. Conditioning was TBI-based in 14 patients and consisted of fludarabine and cyclophosphamide (CY) plus either 12 Gy TBI in all remission patients or 8 Gy TBI in all patients being older than 55 years; all infants (n = 3) received 12 Gy TBI plus etoposide. In adults with relapse after a first allogeneic transplantation conditioning was drug-based: fludarabine, CY plus treosulfan (3 x 10–12g/m2) and etoposide. Post-grafting immunosuppression was high-dose CY, tacrolimus and MMF in all patients. 23/25 patients engrafted, 2 patients died early in aplasia. No primary graft rejection was observed. Acute GvHD grade II-IV occurred in 5 patients (20%), while 6 patients (24%) suffered from mostly mild to moderate chronic GvHD. Severe toxicity (grade III-IV) was observed in 11 patients (44%); most commonly mucositis (36%), transient elevation of transaminases (32%) and diarrhoea (32%). Kidney failure requiring haemodialysis occurred in 3 patients. CMV reactivated in 8 patients and EBV in 3 patients while no patient developed CMV disease or PTLD. Proven invasive aspergillosis was diagnosed in 2 patients. One-year non-relapse mortality was 12%. After a median follow up of 16.6 months, estimated one-year overall survival and relapse-free survival was 74% and 48%, respectively. In summary, intensification of conditioning in the setting of TCR haplo-HSCT using PTCY is well tolerated with low NRM in patients with high-risk and relapsed ALL, while providing an effective anti-leukemic activity in advanced disease. Thus, we suggest that donor availability can be expanded in patients with high-risk and advanced ALL who lack a conventional donor or suffer from aggressive disease. Disclosure: No conflict of interest disclosed. V128

Predictive markers for CD34+ mobilization in healthy peripheral blood stem cell donors Körper S.1, Hauber D.1, Fürst D.1, Reinhardt P.1, Schauweker P.1, Mytilineos J.1, Schwarz K.1, Bunjes D.2, Wiesneth M.1, Schrezenmeier H.1 Universitätsklinik Ulm/DRK Butspendedienst Baden-Württemberg-Hessen, IKT-Ulm, Ulm, Germany, 2Universitätsklinik Ulm, Medizinische Klinik III, Ulm, Germany 1

A few prognostic factors for stem cell mobilization have been established. Body weight (BW) and male sex (MS) are accepted predictors for good mobilization. In search for other prognostic factors we performed a retrospective analysis of the records of 316 healthy stem cell donors.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

35


CONTENTS AUTHOR INDEX

A single platform assay was used to determine the CD34+ cell count/µl in peripheral blood after G-CSF mobilization (aim: 40 µg G-CSF per kg BW, divided in 8 dosages). The concentration of CD34+ cells was correlated with different parameters in all 316 donors. Spearmans Test was used to determine significance (P ≤ 0.01). The study population included 104 women and 212 men. The median age was 38.5 years. The median CD34+ cell concentration after G-CSF was 63/ µl (7 to 280/µl; non-Gaussian distribution). For further analysis groups of poor (PM) and good mobilizers (GM) were defined by the 10. (28/µl) and 90. (132/µl) percentiles. Colony assays differed between PM and GM with BFUE of 49 (18–228) and CFU-GEMM of 10 (4–36) in PM and BFUE of 226 (83–3048) and CFU-GEMM of 47 (16–487) in GM (unit: /105 viable cells). BMI, BW and ALT, and cholinesterase (obtained before G-CSF administration) showed a weak but significant correlation with CD34+ mobilization in men and women. To compare the ultrasound examinations of the liver the presence of steatosis hepatis was examined in the PM and GM. Steatosis of grade 1 or 2 was present in 6% of PM and 41% of GM, a grade 2 steatosis was not seen in PM but in 9% of GM. Further factors were shown in the table. Tab. 1.

Variable

Poor mobilizers (PM) n = 32 Median (range)

Good mobilizers (GM) n = 32 Median (range)

Spearmans Test n = 316 P- value

Weight (kg)

69 (46 to 110)

85 (62 to 140)

<0.0001

Eosinophils (/µl)

109 (17 to 331)

173 (54 to 918)

<0.0001

RBC (*10e6/µl)

4.74 (4.05 to 5.50)

5.28 (4.62 to 6.27)

<0.0001

Reticulocytes (10e3/µl)

54 (34 to 114)

59 (36 to 104)

0.0218

MCV (fl)

87.6 (79.1 to 94.5)

84.9 (79.8 to 90.7)

0.0007

ALT (U/l)

19 (6 to 80)

28 (12 to 79)

<0.0001

AST (U/l)

29 (24 to 58)

32 (20 to 94)

0.0557

Cholinesterase

7176 (4438 to 9881)

8605 (5239 to 12 217)

<0.0001

Liver size (MCL) (mm)

137 (103 to 164)

141 (120 to 178)

0.4428

Our data confirmed MS and BW as positive predictors for CD34+ mobilization. As expected colony growth and stem cell yield was tightly connected to mobilization. Interestingly higher counts of red blood cells and eosinophils (prior to G-CSF) were correlated with better mobilization in an univariate analysis. Indicators of obesity like elevated liver enzymes and steatosis hepatis were also linked to better CD34+ mobilization.

V129

Donor lymphocyte infusion (DLI) in patients with hematological malignancies and the detection of cytotoxic T-cell (CTL) responses against several leukemia-associatedantigens (LAA) Hofmann S.1, Götz M.1, Herbst C.1, Schneider V.1, Wiesneth M.2, Döhner H.1, Bunjes D.1, Greiner J.1 University of Ulm, Department of Internal Medicine III, Ulm, Germany, University of Ulm, Institute of Clinical Transfusion Medicine, Ulm, Germany

1 2

T-cell responses play a central role in maintaining remission and prolonging overall-survival in patients with hematologic malignancies after allo-HSCT and delayed DLI. The role of LAA and mHag has to be elucidated although there is evidence that the graft-versus-leukemia-effect observed after DLI is based on CTL-mediated immunity which is reactive against mHag and LAA. In this study, we analysed peripheral blood and serum samples of 10 patients (pts) with AML (3 pts), CML (2 pts), Multiple Myeloma (3 pts) and CLL (2 pts) in the course of allo-HSCT and before and after DLI for specific CTL responses against several LAA and established a cytokine profile. 3 pts received preemptive DLI and 8 pts received therapeutic DLI. 5 pts underwent a single DLI, 4 pts received a second and 1 pt a third DLI. Epitopes derived from PRAME, NPM1mut, RHAMM, WT-1 and other LAA were tested. Immune reactions of CTL were measured in ELISpot assays (INF-gamma and granzyme B). The corresponding cytokine profile of several cytokines was analysed for further interpretation of the clinical data. In general, 4 of the 10 pts who underwent DLI developed GvHD with 3 pts receiving ongoing immunosuppression. In all patients we could detect a CTL response against at least one of the tested LAA in the course of DLI. The two pts with AML with NPM1-mutation (NPM1mut) who received preemptive DLI in molecular relapse showed a polyspecific CTL-response against several epitopes including those derived from the mutated region of NPM1, Survivin and WT1. CTL responses against RHAMM were detected in AML, Multiple Myeloma, CML and CLL. All tested patients with Multiple Myeloma and CLL showed CTL responses against NYESO1. WT1 was detected in AML and Multiple Myeloma in a high frequency. The pattern of CTL-responses changed during the course, interestingly, patients with GvHD and immunosuppression also showed CTL positivity against several LAA. Immunoinhibitory as well as immunostimulating cytokines were analysed. Cytokine profiles differed in accordance to the clinical situation including immunosuppression, GvHD, time point after DLI application and disease status. Taken together, CTL responses against several LAA were detected in the course of DLI, this might support the hypothesis that these LAAs are a therapeutic target in remission induction in patients with allogeneic transplantation. Patients who developed GvHD after DLI showed a combination of LAA positivity. Disclosure: No conflict of interest disclosed.

Disclosure: Sixten Körper: Employment or Leadership Position: Abteilungsleiter Im IKT Hubert Schrezenmeier: Employment or Leadership Position: Ärztlicher Direktor am IKT; Expert Testimony: Beobachtunrgsstudie zur Anwednung von Filgrastim Hexal.

36

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag

V131

Immuntherapie I

Next-generation dendritic cell vaccination in postremission therapy of AML: results of a clinical phase I trial

V130

Lichtenegger F.S.1,2, Schnorfeil F.M.1,2, Brüggemann M.3, Moosmann A.4, Köhnke T.1, Bücklein V.1, Altmann T.1, Wagner B.5, Hiddemann W.1, Bigalke I.6, Kvalheim G.6, Subklewe M.1,2

Targeting HLA.A2-restricted MDM2 (81–88) epitope as a new antigen for TCR gene therapy of multiple myeloma Amann E.1, Antunes E.1, Jacobi B.1, Theobald M.1,2,3, Echchannaoui H.1,3 University Medical Center of the Johannes Gutenberg University Mainz, Third Medical Department (Hematology, Oncology and Pneumology), Mainz, Germany, 2Johannes Gutenberg University Mainz, Research Center Immunology (FZI), Mainz, Germany, 3German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Frankfurt/Mainz, Germany 1

Background: Adoptive transfer of T cells retrovirally equipped with tumor-associated antigen (TAA)-specific T cell receptor (TCR) has shown efficacy in cancer patients. The human homologous of the murine double-minute 2 protein (MDM2) TAA is overexpressed in a variety of human tumors, including soft tissue sarcoma, multiple myeloma (MM), glioblastoma and melanoma. MDM2 protein overexpression is particularly observed in invasive and metastatic melanoma and is associated with enhanced survival of MM cells. We have generated and optimized an HLA. A2.1-restricted CD8-dependent MDM2 (81–88)-specific TCR isolated from a high-avidity murine CTL clone derived from CD8xA2Kb transgenic mice. Our aim is to target MM in adoptive immunotherapy using human T cells genetically equipped with MDM2-specific TCR. Methods: The nucleotide sequence of the MDM2-TCR-construct was codon-optimized (opt.) and TCR chains cloned in a bicistronic retroviral vector containing the self-cleaving 2A virus-derived peptide. We also introduced an additional inter-chain disulfide bond (cys.) between TCR α and β constant domains to prevent mixed heterodimers formation. Human T cells were retrovirally transduced with TAA-specific TCR and expression levels were analyzed by flow cytometry. HLA.A2.1 MM cell lines were screened for MDM2 expression by Western Blot. In a xenograft mouse model MM cell line was injected subcutaneously in the flank of NOD-scid IL2R gamma chainnull (NSG) mice and human T cells transduced with cys.opt. TCR or mock vector were injected intravenously when the tumors were palpable. Results: We observed a strong correlation between MDM2 expression level and MDM2 TCR-mediated lysis in vitro. In a xenograft mouse model we could observe a prolonged overall survival and tumor-infiltrating T cells in mice which received MDM2-specific TCR transduced T cells compared to Mock-treated group. Interestingly, ex vivo tumor cells exhibit a down-regulation of MDM2 expression and concomitantly an up-regulation of p53 expression which results in a lower recognition of these tumor cells by the MDM2-specific TCR. Preliminary data showed a potent lysis of ex vivo tumor cells by MDM2/p53 dual TCR-modified T cells as compared to single TCR-equipped T cells. Conclusion: Our data show that MDM2-specifc TCR can target MM in vitro and in vivo. Using MDM2- and p53-specific dual TCR transduced T cells may represent a novel approach to circumvent tumor escape mechanisms like antigen down-regulation. Disclosure: No conflict of interest disclosed.

Klinikum der Universität München, Department of Internal Medicine III, Munich, Germany, 2Helmholtz-Zentrum München, Clinical Cooperation Group Immunotherapy, Munich, Germany, 3University Hospital Schleswig-Holstein, Department of Hematology, Kiel, Germany, 4Helmholtz-Zentrum München, Clinical Cooperation Group Immunooncology, Munich, Germany, 5Klinikum der Universität München, Department of Transfusion Medicine, Cellular Therapeutics and Hemostaseology, Munich, Germany, 6The Norwegian Radium Hospital, Oslo University Hospital, Department of Cellular Therapy, Oslo, Norway 1

Introduction: Postremission therapy for acute myeloid leukemia (AML) is critical for elimination of minimal residual disease (MRD). In patients not eligible for allogeneic stem cell transplantation, therapeutic vaccination with autologous, tumor antigen-loaded dendritic cells (DCs) is a promising treatment strategy to induce anti-leukemic immune responses and to eradicate chemorefractory cells. We have developed a GMP-compliant 3-day protocol including a TLR7/8 agonist to differentiate monocytes of intensively pretreated AML patients into next-generation DCs. Methods: We are conducting a proof-of-concept phase I/II (6+14 patients) clinical trial using next-generation DCs as postremission therapy for AML of non-favorable genetic risk in CR after intensive induction therapy (NCT01734304). DCs are loaded with RNA encoding the leukemia-associated antigens WT1 and PRAME as well as CMVpp65 as adjuvant and surrogate antigen. Patients are vaccinated intradermally with 5×106 DCs of each antigen species up to 10 times within 26 weeks. The primary endpoint of the trial is feasibility and safety of the vaccination. Secondary endpoints are immunological responses and disease control. Results: 9 patients have been enrolled into the trial. With one dropout, DCs of sufficient number and quality were generated from leukapheresis in 7/8 cases. DC analysis showed a positive costimulatory profile, secretion of IL-12p70, migration towards a chemokine gradient, antigen expression and specific T cell activation in vitro. 4 patients have completed the vaccination schedule; the 5th patient has received 7/10 vaccinations. We observed delayed-type hypersensitivity (DTH) responses at the vaccination site in 5/5 patients, accompanied by slight erythema and indurations at the injection site, but no grade III/IV toxicities. Multimer analysis revealed the induction of antigen-specific T cell responses in 3/3 patients tested. We detected an increase of WT1-specific T cells in one patient and strong inductions of CMVpp65-specific T cells in two CMV-seronegative patients. TCR repertoire analysis by next-generation sequencing revealed an enrichment of particular clonotypes at DTH sites. In an individual treatment attempt, we treated one patient with impending relapse with a combination of DC vaccination and 5-azacytidine, resulting in MRD conversion. Conclusion: Vaccination with next-generation DCs in AML is feasible and safe, and induces anti-leukemia-specific immune responses in vivo. Disclosure: No conflict of interest disclosed. V132

Selective engagement of the RIG-I pathway synergizes with checkpoint blockade in cancer immunotherapy Heidegger S.1, Kreppel D.1, Bscheider M.1,2, Wintges A.1, Bek S.1, Schmickl M.1, Fischer J.C.1, Lin C.-C.1, Peschel C.1, Haas T.1, Poeck H.1,3 Klinikum rechts der Isar der Technischen Universität München, III. Medizinische Klinik, München, Germany, 2Stanford University School of Medicine, Department of Pathology, Stanford, United States, 3Memorial Sloan-Kettering Cancer Center, Departments of Immunology and Medicine, and Cell Biology, New York, United States 1

Introduction: Targeting of regulatory T cell receptors such as CTLA-4 has been shown to enhance anti-tumor immune responses. Given that

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

37


CONTENTS AUTHOR INDEX

checkpoint inhibitors can augment T-cell reactivity against tumor neoantigens, we aimed at improving this response with a vaccination protocol that combines selective engagement of the nucleic acid-sensing pattern recognition receptor RIG-I, antigen and CTLA-4 blockade. Methods: We used wild-type and genetically-altered mice as well as an established combination of antibiotics to characterize the molecular pathways and external influences affecting our combinatorial approach on a whole organism level. Specifically, mice were vaccinated with ovalbumin (OVA) and the specific RIG-I ligand 5’-triphosphat RNA (3pRNA) together with anti-CTLA-4 antibody. The frequency of OVA-specific T cells and specific lysis of target cells was determined. For tumor challenge, identically treated mice were injected iv with B16.OVA melanoma cells and the number of lung metastases was analyzed. By using 3pRNA/ OVA-stimulated dendritic cells (DCs) isolated from gene-deficient mice, we assessed the relevance of these respective pathways for cross-presentation and subsequent cross-priming of co-cultured cytotoxic T cells. Results: We found that vaccination and RIG-I ligation strongly synergized with CTLA-4 blockade to induce expansion and activation of antigen-specific CD8+ T cells and potent anti-tumor immunity (Figure 1). Cross-priming of cytotoxic T cells as well as anti-tumor immunity required the adapter protein MAVS and type I interferon (IFN) signaling and were mediated by DCs. In addition, the benefit of the combined immunization with anti-CTLA-4 was reduced by systemic antibiotics pointing to the prerequisite of an intact commensal microbiota in this context. Conclusion: We established a novel combinatorial strategy to generate peptide-specific CD8+ T cells and anti-tumor immunity and characterized the respective signaling pathways, cell types and environmental influences responsible for this beneficial response. The combination of RIG-I-induced type I IFN and checkpoint blockade has the potential to enhance personalized anti-cancer vaccines.

tumor-specific, the elimination of healthy non-cancerous cells may occur. To improve tumor cell-selectivity we envisioned a “dual-targeting” approach: Antibody derivatives in the triplebody format simultaneously target two different TAAs on the surface of the same tumor cell and engage immune effector cells such as NK or T cells via a trigger antigen. Methods: Triplebody 33-3-19 was constructed from single chain variable fragment (scFv) building blocks, expressed recombinantly in suspension-adapted FreeStyle™ 293-F cells and purified. Protein properties and cytotoxic activity were analyzed using molecular biology and flow cytometric techniques as well as redirected lysis assays. Results: 33-3-19 binds the B lymphoid marker CD19 and the myeloid marker CD33 specifically; an antigen combination that is exclusively present on biphenotypic leukemia cells. Furthermore, it recruits T cells via the CD3 epsilon chain and activates these effectors as efficiently as the recently described mono-targeting triplebody 19–3-19 (Roskopf et al. 2014). The triplebody induced specific lysis of established myeloid (MOLM13, THP-1) and B-lymphoid cell lines (SEM, Raji) and of primary patient cells at low picomolar concentrations. EC50 values ranged from 0.1 to 28 pM. At 1 nM concentration 33–3-19 induced preferential lysis of double- over single-positive leukemia cells at a ratio of 2-to-1 in a target cell mixture: (CD19 plus CD33) double-positive BV173 target cells were eliminated more efficiently in the presence of 33–3-19 plus T cells than the CD19 single-positive SEM target cells, in spite of comparable overall target antigen densities (50,000–60,000/cell). 19–3-19 or a bispecific T cell engager mixture of 19–3 and 33–3, however, led to similar elimination of both cell lines at 1nM. Conclusions: Dual-targeting agents such as triplebodies may be used for an efficient and more selective immune-intervention in cancer and may induce the elimination of leukemia-initiating cells. Disclosure: No conflict of interest disclosed. V134

Combining immune checkpoint inhibition with the CD33 BiTE® antibody construct AMG 330 to enhance T cell mediated lysis of primary AML cells Krupka C.1,2, Kufer P.3, Kischel R.3, Zugmaier G.3, Köhnke T.1,2, Lichtenegger F.S.1,2, Altmann T.1,2, Spiekermann K.1,4, Vick B.4,5, Jeremias I.4,5, Hiddemann W.1,4, Subklewe M.1,2,4 Klinikum der Universität München, Department of Internal Medicine III, München, Germany, 2Helmholtz Institute Munich, Clinical Co-operation Group Immunotherapy, München, Germany, 3AMGEN Research (Munich) GmbH, München, Germany, 4German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany, 5Helmholtz Zentrum München, Department of Gene Vectors, München, Germany 1

Fig. 1. Disclosure: No conflict of interest disclosed. V133

Triplebody 33-3-19 eliminates biphenotypic (CD19 plus CD33) leukemia cells selectively Roskopf C.C.1, Braciak T.A.1, Fenn N.2, Kobold S.3, Jacob U.4, Fey G.H.4, Hopfner K.-P.2, Oduncu F.S.1 Klinikum der Universität München, Medizinische Klinik und Poliklinik IV, Hämatologie/Onkologie, München, Germany, 2Genzentrum, München, Germany, 3Klinikum der Universität München, Medizinische Klinik und Poliklinik IV, Klinische Pharmakologie, München, Germany, 4SpectraMab GmbH, München, Germany 1

Introduction: Targeted immune-interventions with antibodies and antibody derivatives have shown improved tumor-cell specificity for cancer therapy. However, as target antigens are tumor-associated (TAA) but not

38

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: BiTE® antibody constructs are very efficient in inducing T-cell activation and secretion of proinflammatory cytokines (Krupka, Blood 2014). The proinflammatory cytokine milieu favors the upregulation of immune checkpoint molecules on target cells. In this study the relevance of immune checkpoint molecules in CD33/CD3 BiTE® antibody construct (AMG 330) mediated lysis of primary AML cells was evaluated. Methods: AMG 330 mediated lysis of primary AML cells was analyzed in a long-term culture system. The assay system was validated for proliferation of AML cells, antigen expression pattern, clonal evolution and persistence of leukemia initiating cells (LIC). The expression level of checkpoint molecules was assessed by flow cytometry (MFI ratio). Results: Ex vivo analysis of primary AML cells in long-term cultures showed consistent AML cell proliferation or persistence (n = 38), stable antigen expression pattern (n = 19) and clonal stability (n = 2) throughout at least 28 days of culture. In vivo xenotransplantation experiments confirmed the propagation of LICs in the system (n = 4). Although not constitutively expressed on primary AML cells (n = 123), PD-L1 was strongly upregulated upon the addition of AMG 330 to the ex vivo cytotoxicity experiments (p < 0.0001; n = 27). This phenomenon was

Abstracts


CONTENTS AUTHOR INDEX

cytokine-mediated as the sole addition of IFN-γ and TNF-α also induced expression (n = 6). Furthermore, we observed a significant upregualtion of PD-1 on activated T cells (MFI ratio: control vs AMG 330 3.1 : 12.9, p = 0.0002, n = 18). In ex vivo cytotoxicity experiments blocking of the PD-1/PD-L1 interaction significantly enhanced AMG 330 mediated lysis efficacy (median lysis: AMG 330 58% vs + α-PD-1/PD-L1 75%, n = 9, p = 0.03) which was accompanied by a significant increase in T cell proliferation and a markedly increase in IFN-γ secretion. The effect was most prominent in cultures with low E:T ratios (PD-1: lysis E:T 1:1: AMG 330 100% vs + α-PD-1 100%; lysis E:T 1:9: AMG 330 90% vs + α-PD-1 98%; PD-L1: lysis E:T 1:1: AMG 330 95% vs + α-PD-L1 100;% lysis E:T 1:5: AMG 330 83% vs + α-PD-L1 97%). Our work thus provides evidence that PD-L1 upregulation is a relevant mechanism of primary AML cells to escape cytokine-mediated immune responses. We demonstrate that AMG 330 mediated cytotoxicity is enhanced by blockade of inhibitory receptors on AML cells. Conclusion: Our results support the use of combinatorial approaches of BiTE® antibodies with immune checkpoint blockade. Disclosure: Christina Krupka: Expert Testimony: Die Studie wurde teilweise durch AMGEN Research (Munich) finanziert Marion Subklewe: Expert Testimony: Die Studie wurde teilweise durch AMGEN Research (Munich) finanziert. V135

Modulation of glycolytic metabolism promotes in vitro generation of EBV-reactive and redirected virus-specific human CD8+ cytotoxic T lymphocytes with stem-cell-memory and central-memory properties Weber I.1, Bhatti A.1, Krebs L.1, Theobald M.1, Hartwig U.F.1 III. Medizinische Klinik, Universitätsmedizin der Johannes GutenbergUniversität Mainz, Hämatologie, Internistische Onkologie & Pneumologie, Mainz, Germany 1

Introduction: Adoptive T cell therapy has advanced as a powerful tool to treat opportunistic viral infections and leukemia relapse. However, terminally differentiated, high avidity effector T cells (TEFF) are often limited to establish durable responses whereas less differentiated stem-cell-memory (TSCM) and central-memory (TCM) T cells can elicit potent antitumor immunity, prolonged survival and memory. Moreover, TSCM and TCM depend less on glucose consumption to drive oxidative phosphorylation (OXPHOS) as their primary source of ATP whereas TEFF utilize aerobic glycolysis to generate additional ATP. We therefore studied means of modulating T cell metabolism to generate EBV-specific TSCM and TCM for adoptive transfer and, additionally, for redirecting robust viral specificities to leukemia by e.g. retroviral T cell receptor (TCR)-transfer. Methods: Naive HLA-A2+CD8+CD45RA+ T cells and total CD8+ T cells were stimulated by autologous DCs and subsequently PBMCs loaded with EBV-peptides in the presence of low glucose (1 mM), glutamine, an optimized cytokine cocktail and 1 mM of either glucose analog 2-deoxy-glucose (2-DG), galactose or 9-oleic acid. In addition to phenotypic and functional analyses glucose uptake and lactate production was determined. OXPHOS and aerobic glycolysis was assessed by measuring oxygen-consumption rate (OCR) and extracellular acidification rate (ECAR) on a Seahorse Analyzer. Results: Repetitive restimulation of naive T cells revealed EBV-reactive CTL expressing a CD8+CD45RA+CD45RO- TSCM and CD8+CD45RACD45RO+ TCM phenotype in the presence of low glucose, glutamine and 2-DG when compared to untreated CTL. This effect was also seen in total CD8+ T cells although less pronounced. In contrast, supply of galactose or oleic-acid to glucose free (but glutamine containing) medium did not result in reduced TEFF differentiation, suggesting OXPHOS by galactose metabolism and fatty acid oxidation. Moreover, 2-DG treated TSCM and TCM showed less lactate production and ECAR as compared to untreated controls, suggesting that differentiation to TEFF requires aerobic glycolytic ATP production. 2-DG treated TSCM and TCM revealed EBV-reactivity

40

Oncol Res Treat 2015;38(suppl 5):1–270

comparable to controls, elicited superior migration properties in vitro and could be sucessfully reprogrammed with an AML-reactive TCR. Conclusions: These studies demonstrate that modulating T cell metabolism may be a promising approach to generate TSCM/CM in vitro for improved adoptive cellular therapy. Disclosure: No conflict of interest disclosed.

Expertenseminar Genetische Beratung V137

Genetic counselling Müller H.1 Universitätsspital Basel, Medizinische Genetik, Basel, Switzerland

1

Genetic counselling is a communication process between patients or persons at risk of a disorder that may be hereditary and health care professionals. It seeks to assist the counselees to understand medical and genetic facts of the disorders. However it does not stop with informing about those facts, but must include a variety of other actions if it is to be effective. Genetic counselling gains practical importance in haematology and oncology due to the progress of molecular genetic testing. The following topics will be discussed: – The general concept of genetic counselling (diagnostic aspects, estimation of risks, supportive role) and its realization in daily practice, – Genetic counselling in Mendelian and in common disorders, – Counselling of already affected individuals, – Genetic risk estimation for healthy family members, – Ways of communication and empathy for counselees, – Genetic testing and counselling of children and adolescent, – Inclusion of healthy family members (related and unrelated), – Autonomy of the counselee, shared decision making, non-directiveness, informed consent, – Gene- versus gene panel-sequencing, – Handling of an uncertain or even wrong diagnosis, – Interdisciplinary collaboration between the medical specialists involved/the role of case managers, – Perception of consanguinity and ethnicity, – Sources for current scientific information concerning genetic disorders, – Legal aspects concerning genetic testing and counselling, – Teratogenic and genetic consequences of chemotherapy and radiation. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium

Kolon- und Rektumkarzinom Optimierungen der Strategien V143

Are less intensive/sequential therapies in elderly and fragile patients with mCRC are justified? Eisterer W.1 Medizinische Universität Innsbruck, Innere Medizin V, Innsbruck, Austria

1

Colorectal cancer has a high prevalence in the aging population, with a median age of 69 years at diagnosis. The vast majority of patients with colon cancer will require chemotherapy treatments during their disease course, challenging oncologists with the task of tailoring therapy for older patients with CRC in the face of limited evidence-based data to guide them. Older patients, particularly those with a higher numberof comorbidities, are less likely to be referred to a medical oncologist after diagnosis and once referred, they have a lower likelihoodof receiving chemotherapy. Recent Phase-III studies (1–3) showed no inferior overall survival (OS) if

Abstracts


CONTENTS AUTHOR INDEX

5-fluorouracil (5FU) or capecitabine was chosen as first-line therapy versus combination chemotherapy. The MRC FOCUS2 trial (3) was conducted specifically among older and frail patients with untreated mCRC. Patients were randomly assigned to receive capecitabine or 5FU, with or without oxaliplatin, at a 20% dose reduction. Capecitabine and 5FU showed similar clinical benefit in progressions-free survival (PFS) and OS. The addition of Oxaliplatin with a 20% dose reduction resulted in improved Response rate and a lack of improvement in OS. The randomized phase III AVEX Trial (4) compared capecitabine alone or in combination with bevacizumab and enrolled untreated patients 70 years or older who were not candidates for oxaliplatin or irinotecan. AVEX demonstrated prolonged PFS, better response and a trend toward improved OS. Conclusion: Less intensive/sequential therapies are justified in elderly and/or fragile patients with mCRC. References: 1 Seymour MT et al. Chemotherapy options in elderly and frail patients with metastatic colorectal cancer (MRC FOCUS2): an open-label, randomised factorial trial. Lancet 377:1749–1759,2011. 2 Koopman M et al. Sequential versus combination chemotherapy with capecitabine, irinotecan, and oxaliplatin in advanced colorectal cancer (CAIRO): A phase III randomised controlled trial. Lancet 370:135–142,2007. 3 Ducreux M et al.: Sequential versus combination chemotherapy for the treatment of advanced colorectal cancer (FFCD 2000–05): An open-label, randomised, phase 3 trial. Lancet Oncol 12:1032–1044,2011. 4 Cunningham D, et al.: Bevacizumab plus capecitabine versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer (AVEX): an open-label, randomized phase 3 trial. Lancet Oncol 14:1077–85, 2013. Disclosure: No conflict of interest disclosed.

Fortbildung

HIV-assoziierte Tumoren V147

Aggressive HIV-associated B-cell lymphoma: options and limits of therapeutic approaches Hübel K.1 Universitätsklinik Köln, Klinik I für Innere Medizin, Köln, Germany

1

Patients infected with HIV have an increased risk of developing aggressive B-cell non-Hodgkin’s lymphoma (NHL). Recent data indicate that among AIDS-related death NHL is the most frequent event. The majority of patients have poor prognostic features such as advanced-stage disease or high International Prognostic Index (IPI). Since the introduction of combination antiretroviral therapy (cART) therapeutic options and prognosis have considerable improved. However, the overall outcome remains worse compared to immunocompetent patients. Work-up and staging of patients should be performed in accordance with guidelines established for HIV-negative NHL. In diffuse large B-cell lymphoma (DLBCL), most centers use 6 cycles of R-CHOP 21 as first-line treatment achieving a complete remission in about 60% of patients. Date clearly show that the addition of rituximab improves outcome compared to CHOP alone. However, if CD4 cell counts is below 50/µl, rituximab is not recommended due to increased risk of infection. The use of R-EPOCH may further improve prognosis compared to R-CHOP. In Germany, a prospective trial evaluating the R-CHOEP regimen as first-line treatment in DLBCL will be initiated soon. In the relapse situation, patients may be candidates for high-dose therapy followed by autologous stem cell transplantation as in the HIV-negative setting. In Burkitt lymphoma, does-intensive regimens such as the B-ALL/NHL2002 protocol of the GMALL or the Hyper-CVAD/HD-MTX protocol are successfully applied to patients who are in an acceptable condition. Patients with rare, aggressive diseases as plasmablastic lymphoma or primary effusion lymphoma represent

Abstracts

a therapeutic challenge for clinicians since no therapeutic approach has been clearly recommended yet. There is no doubt the cART applied concurrently to chemotherapy is an essential module to a successful therapeutic outcome provided that pharmacokinetik interactions are being considered. In summary, significant advances have been made in the understanding and treatment of HIV-associated B-cell lymphomas in recent years. Prospective trials are needed to develop treatment algorithms and to investigate novel therapeutic approaches for patients with HIV-lymphoma. Disclosure: No conflict of interest disclosed. V148

Non-AIDS-defining malignancies: current therapeutic concepts Hentrich M.1 Rotkreuzklinikum, München, Germany

1

Combined antiretroviral therapy (cART) has dramatically improved the life expectancy of patients living with HIV. However, the risk of cancer is substantially increased in HIV-infected patients. In western countries the number of cases of non-AIDS-defining malignancies (NADM) now equals or exceeds the number of cases of AIDS-defining cancers, and NADM are a leading cause of mortality for HIV-infected subjects. Important risk factors for NADM are advanced age, the length of time one has been infected with HIV, exposure to cigarette smoking, and oncogenic viruses such as Epstein-Barr virus, Hepatitis B and C virus, and human papillomavirus. The most frequent NADM are lung cancer (LC), Hodgkin lymphoma (HL), anal cancer (AC), liver cancer (HCC) and head & neck cancer. Basically, patients with NADM should be treated in an identical manner to patients without HIV infection. Most cases of LC occur in the setting of limited immune deficiency and a long-lasting viral suppression. However, as in the HIV-negative setting long-term overall survival can only be achieved at limited LC stages. Improved survival rates were reported for patients undergoing stage-adapted treatment of HIV-HL. Two cycles of ABVD followed by involved-field (IF) radiation therapy (RT) can be regarded as standard treatment for early favorable HL. Four cycles of ABVD followed by IF-RT are standard of care for early stage unfavourable HL, and patients with advanced stage HL should receive 6 cycles of BEACOPP baseline or 6–8 cycles of ABVD. The management of AC with chemoradiotherapy (CRT) achieves similar outcomes as the general population. However, CRT is associated with significant prolonged CD4 suppression. The outcome of HCC in the setting of HIV remains poor. During radio- or chemotherapy patients should receive concurrent cART. However, interactions between cytotoxics and antiretrovirals must be considered, as chemotherapy-related toxicity may be markedly increased by concomitant use of antiretrovirals. Antimicrobial prophylaxis should be considered in accordance with current guidelines. In particular, prophylaxis against pneumocystis jiroveci pneumonia is indicated if the CD4 cells are <200/µl. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium Maligne Gliome V152

Recent diagnostics, therapy standards and perspectives in patients with anaplastic gliomas and glioblastomas (WHO grade III/IV) Hau P.1 Universität Regensburg, Neurologie, Regensburg, Germany

1

Anaplastic gliomas and glioblastomas cannot be cured at this time. However, genetic markers increasingly enable prognostication and allow strat-

Oncol Res Treat 2015;38(suppl 5):1–270

41


CONTENTS AUTHOR INDEX

ification into specific treatment groups, even if the standard histology is not distinctive. The most widely used drug in the treatment of gliomas is the alkylating agent Temozolomide. In addition, other alkylating agents and the antiangiogenic substance Bevacizumab are used. Bevacizumab generates impressive efficacy in a subgroup of patients with gliomas. However, after two negative Phase III-trials, the substance is not approved in all countries. Immunotherapeutic strategies become increasingly interesting. Relevant, beyond others, are the induction of humoral and celluar immune responses against EGFRvIII and the mutated form of IDH1 (IDH1R132H), autologous dendritic cell vaccinations with e.g. peptide cocktails (ICT-107), and use of immune checkpoint blocking agents against PD-1/PD-L1 and CTLA-4. A novel concept uses tumor treating fields (TTF, Optune) that induce apoptosis. A final decision on this treatment can be made after full publication of a completed Phase III-trial that is expected this summer. Therapeutic decisions increasingly include genetic markers. The methylation of the promotor of O6-Methyl-Guanin-Methyl-Transferase (MGMT) and the codeletion of chromosome arms 1p and 19q (LOH1p1/19q) have been evaluated as predictive markers in a number of trials. They are routinely used in at least two situations for treatment stratification, namely in elderly patients with glioblastoma (where MGMT is used to stratify to radiotherapy or chemotherapy) and in patients with anaplastic glioma with 1p19q codeletion. In these patients, a combination of radiotherapy and chemotherapy with Procarbacin, CCNU and Vincristin doubles overall survival in comparison to radiotherapy as monotherapy. The development in the use of genetic markers for prognostication and therapy stratification is dynamic. Considering this, a new classification of gliomas will soon include genetic markers and will therefore more thoroughly inform the treating physician about the best treatment of choice. Disclosure: No conflict of interest disclosed. V153

Recommendations for the treatment of recurrent malignant glioma Weller M.1 Universitätsspital Zürich, Klinik für Neurologie, Zürich, Switzerland

1

Newly diagnosed glioblastoma is now commonly treated with surgery as feasible or biopsy, followed by radiotherapy plus concomitant and adjuvant temozolomide. The treatment of recurrent glioblastoma is less well standardized and depends on multiple factors, including efficacy and tolerability of prior treatment, time from diagosis, topographic pattern of relapse, and MGMT promoter methylation status. A minority of patients are candidates for second surgery or reirradiation, based on tumor size and location. Progression-free survival rates at 6 months of 20%-30% have been reported with nitrosoureas, temozolomide rechallenge in various dosing regimens, or bevacizumab. Temozolomide rechallenge is not active in patients with tumors lacking MGMT promoter methylation (DIRECTOR) whereas bevacizumab is not approved in the EU. Data from a phase III trial (EORTC 26101) comparing the combination of lomustin and bevacizumab versus lomustin alone are expected near the end of the year. Emerging therapies may include vaccination against mutant epidermal growth factor receptor variant III (EGFRvIII) (ReACT) and the PD-1 antibody nivolumab. More research is needed to better define patient profiles that predict benefit from the limited therapeutic options available after the current standard of care has failed. Disclosure: Michael Weller: Financing of Scientific Research: Celldex, Immunocellular, Isarna, Magforce, MSD, Merck & Co, Northwest Biotherapeutics, Novocure, Pfizer, Roche, Teva; Expert Testimony: Acceleron, Actelion, Alpinia Institute, Bayer, Isarna, MSD, Merck & Co, Novocure, PIQUR, Roche.

Fortbildung

Kopf-, Hals-, Schild- und Speicheldrüsentumore – Update 2015 V154

Thyroid cancers: which molecular marker are relevant? Siano M.1,2 Cantonal Hospital St. Gallen, Clinic of Oncology / Hematology, St. Gallen, Switzerland, 2Fondazione IRCCS Istituto Nazionale dei Tumori, Head and Neck Medical Oncology Unit, Milan, Italy 1

Even if rarely encountered by the medical oncologist, his knowledge is required for iodine refractory differentiated thyroid cancer (DTC), poorly differentiated and anaplastic TC. In de-differentiated carcinomas as in iodine refractory, molecular alterations play an important role. There is a ‚career of dedifferentiation’ according to mutational status (Xing 2013). Moreover, mutations have direct impact on iodine uptake and so on effectiveness of radioiodine therapy. With targeted therapies e.g. it is possible to convert iodine refractoriness in follicular TC (Hu 2013, Rothenberg 2015). This shows that even if molecular markers are not yet established in decision-making, they will gain importance as prognostic and predictive markers. Mutational landscape was soundly analyzed particularly for papillary TC (Agrawal 2014). Different gene signatures were identified (BRAF-like und RAS-like). Affinity towards specific targets, tumor pathways and gene expression signatures lead to possible assumptions about best treatment or best use of novel tyrosine kinase inhibitors (TKI). Even if tempting, this approach is for the moment, not supported by data. Beside mutational status, angiogenesis seems to be of major importance. TKI with anti-angiogenetic properties, show to be very active (Lenvatinib, Axitinib). This confirms that some genetic alterations are not drivers and that TC serves more mechanisms. Prognostic value of BRAF mutation, even if controversially discussed at primary diagnosis, is associated with higher recurrence rates (Xing 2015) and seems prognostic in advanced disease (in iodine refractoriness). TERT mutation indicates poor prognosis, notably in papillary TC and seems to be associated with distant metastasis and increased disease specific mortality (Melo 2014, Gandolfi 2015). For DTC Sorafenib and Lenvatinib and for medullary TC Vandetanib and Cabozantinib are or will soon be registered. Still these agents are not applied in a selected patient population. BRAF mutational status is not predictive for better outcome with Sorafenib treatment (Brose 2014). Vemurafenib und Dabrafenib are investigated in selected patients harboring a BRAF mutation showing satisfactory response rates. No predictive molecular marker has been identified or was soundly validated so far. Only for Cabozantinib treatment in medullary TC, RET M918T mutation was able to show in a retrospective analysis, predictive potential for overall survival (Elisei 2013, Schlumberger 2015). Disclosure: Marco Siano: Advisory Role: Beratertätigkeit: Bayer, Eisai.

Fortbildung

Ethik Advance Care Planning V159

“Existential communication” at the end of life – conceptual and practical aspects Gieseler F.1, Schäfer V.1, Theobald W.2 UKSH Campus Lübeck, Med I, Palliativmedizin und Ethik in der Onkologie, Lübeck, Germany, 2Agrar- und Ernährungswissenschaftliche Fakultät, Ethik in den Lebenswissenschaften, Kiel, Germany 1

Empathic patient-centered communication is an integral part of the therapeutic care in palliative oncology. The integration in the German National Cancer Plan and the Patients´ Rights Act underlines this consent. Thus,

42

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

the doctor is obliged to explain in an understandable way, all significant information for the diagnosis, all treatment options, and probable side effects (SGB V § 630 c BGB). The requirement is that all service providers have the necessary communication skills (NKP Ziel 12a). These objectives and demands do not consider that terms and conditions in palliative oncology have changed considerably including external environment (space, time frame, etc.), the background of the patient (family, friends, counselors) and the patients themselves (age structure, level and sources of information). We believe that a communication style should be developed, taking account of these changing conditions and thus leading to more patient satisfaction and less “burn-out symptoms” among physicians. Despite the developments of modern oncology, the existential threat by the cancer diagnosis is still vivid. Also doctors and nurses have to cope with finiteness and limitations, e.g. on the time available, treatment options and financial resources. We work on the subject with in the “Projektkolleg Erfahrung und Umgang mit Endlichkeit” of the Collegium Philosophicum at the CAU Kiel. The term of our subproject “Existenzielle Kommunikation” we have borrowed from the philosophy of Carl Jaspers, but augmenting it with the aspect of communication in a situation of existential threat. We found that ethical aspects and communication style represent stressors for patients and physicians alike. One hypothesis is that the communication style is a part of the treatment concept and should be adjusted to the specific situation. Styles include the patriarchal approach, shared decision making and patient centered communication in the sense of service oriented approach. Advance care planning needs to be integrated into the concept. We present here among others a didactic method in teaching communication (“double reflection”). References: Birninger, Gieseler. “Communication between nurses and patients in oncology” Oncol Res Treat 37,2014;S1:110. Mannhardt, Ogbonnaya, Gieseler. “Double Reflection” Oncologist 18 (2013):1058. Gieseler, Schäfer, Theobald “Entscheidungen im Schatten der Endlichkeit “. In “Endlichkeit“, Bihrer (Hrg.), transcript-Verl, accepted. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Allogene Transplantation klinisch I V161

Ruxolitinib for corticosteroid-refractory graft-versus-host disease: analysis of 95 patients treated at multiple medical centers Zeiser R.1, Burchert A.2, Lengerke C.3, Verbeek M.4, Maas-Bauer K.1, Metzelder S.2, Spoerl S.4, Ditschkowski M.5, Ecsedi M.3, Sockel K.6, Ayuk F.7, Ajib S.8, Sicre de Fontbrune F.9, Na I.-K.10, Penter L.10, Holtick U.11, Wolf D.12, Schuler E.13, Meyer E.14, Apostolova P.1, Bertz H.1, Marks R.1, Lübbert M.1, Wäsch R.1, Scheid C.11, Ordemann R.6, Bug G.8, Kobbe G.13, Negrin R.14, Brune M.15, Spyridonidis A.16, Schmitt-Gräff A.17, van der Velden W.18, Huls G.18, Grigoleit G.U.19, Kuball J.20, Blazar B.R.21, Arnold R.10, Kröger N.7, Passweg J.3, Halter J.3, Socié G.22, Beelen D.5, Peschel C.4, Neubauer A.2, Finke J.1, Duyster J.1, von Bubnoff N.1 Freiburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 2Marburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Marburg, Germany, 3University Hospital Basel, Hematology, Basel, Switzerland, 4 Technical University of Munich, III Department of Internal Medicine, Munich, Germany, 5West German Cancer Center, University Hospital Essen, Department of Bone Marrow Transplantation, Essen, Germany, 6Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Department of Hematology, Oncology and Stem Cell Transplantation, Dresden, Germany, 7 University Hospital Hamburg-Eppendorf, Department of Hematology, Oncology and Stem Cell Transplantation, Hamburg, Germany, 8University Hospital Frankfurt/Main, Frankfurt, Germany, 9Saint Louis Hospital, APHP, Hematology Stem cell transplant Unit, Paris, France, 10Charité University Medicine Berlin, Department of Stem Cell Transplantation, Berlin, Germany, 11 University Hospital Cologne, Department of Internal Medicine I, Cologne, Germany, 12University Hospital Bonn (UKB), Medical Clinic III, Oncology, Hematology and Rheumatology, Bonn, Germany, 13Universitätsklinikum Düsseldorf, Department of Hematology, Oncology and Clinical Immunology, Düsseldorf, Germany, 14Stanford University Medical School, Department of Bone Marrow Transplantation, Stanford, United States, 15University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden, 16Patras University Medical School, Department of Bone Marrow Transplantation, Patras, Greece, 17 Freiburg University Medical Center, Department of Pathology, Freiburg, Germany, 18Radboud University Medical Centre, Department of Hematology, Oncology and Stem Cell Transplantation, Nijmegen, Netherlands, 19University Medical Centre Würzburg, Department of Hematology, Oncology and Stem Cell Transplantation, Würzburg, Germany, 20University Medical Center Utrecht, Department of Hematology, Utrecht, Netherlands, 21University of Minnesota, Minneapolis, Division of Blood and Marrow Transplantation, Minnesota, United States, 22Saint Louis Hospital, APHP, Paris, Hematology Stem cell transplant Unit, Paris, France 1

Introduction: Despite major improvements in allogeneic hematopoietic cell transplantation (allo-HCT) over the last decades, severe acute and chronic graft-versus-host disease (GvHD) still remains a life-threatening complication associated with high mortality. Pre-clinical evidence has indicated an anti-inflammatory effect of ruxolitinib, therefore we analyzed responses to ruxolitinib as salvage treatment in patients suffering from corticosteroid refractory GvHD. Methods: A total of 19 Stem Cell Transplant Centers in Europe and United States reported outcome data from 95 patients who received ruxolitinib for corticosteroid-refractory GvHD (skin, mucosa, intestine, liver, lung, musculoskeletal) between 01/2012 and 04/2015. Patients were classified as having acute (n = 54, only grade 3 or 4) or chronic (n = 41, only moderate or severe) GvHD. The median number of previous GvHD-therapies was 3 for acute GvHD (range: 1–7) and 3 for chronic GvHD (range: 1–10). Results: The overall response rate was 81.4% (44/54) in acute GvHD comprising 25 CRs (46%). In chronic GvHD the overall response rate was 85% (35/41). Clinical improvement was at a median time to response of 1.5 (1–10) weeks and 3 (1–25) weeks after initiation of ruxolitinib treatment in acute and chronic GvHD, respectively. GvHD relapsed in 6.8% (3/44) and 5.7% (2/35) of the ruxolitinib-responsive patients with acute or chronic GvHD, respectively. The median follow-up was 22 (3–98) and 25

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

43


CONTENTS AUTHOR INDEX

(2–112) weeks for acute or chronic GvHD patients. Cytopenias (anemia, leukopenia or thrombocytopenia) and CMV reactivation were observed during the time of ruxolitinib treatment in both acute (30/54, 55.5% and 18/54, 33.3%) or chronic (7/41, 17% and 6/41, 14.6%) GvHD patients. Cytopenias had preceded ruxolitinib treatment in 51.8% (28/54) and 14.6% (6/41) of the patients with acute or chronic GvHD, respectively. Relapse of the underlying malignancy occurred in 9.2% (5/54) and 2.4% (1/41) of the patients with acute or chronic GvHD, respectively. Conclusions: Overall, these data collected in multiple centers, using different strategies for GvHD prophylaxis and treatment, suggest that ruxolitinib is a very promising agent in the treatment of corticosteroid-refractory acute or chronic GvHD and may be successfully used to salvage a major subset of patients beyond 2nd line of GvHD treatment and needs to be validated in a prospective trial. Disclosure: No conflict of interest disclosed. V162

Genetic polymorphism of cytochrome P450 1B1 (C432G) is associated with lower overall survival in male patients undergoing allogeneic haematopoietic stem cell transplantation Stute N.1, Beelen D.1, Koldehoff M.1 Clinic for Bone Marrow Transplants, University of Duisburg-Essen, Essen, Germany

1

Introduction: Human cytochrome P450 1B1 (CYP 1B1) is a key enzyme involved in estrogen/drug metabolism, steroid synthesis and pro-carcinogen activation. It is highly expressed in some human cancers and some myeloid cells but not in liver or kidney. The CYP 1B1 codon 432 polymorphism (one of four identified SNP) leads, among other things, to a fourfold higher km in the 4-hydroxylation of 17ß-estradiol. Methods: In a single center retrospective study 382 patients (53% male) who underwent allogeneic HSCT (95% myeloablative) for various diseases (51% acute leukemia) and their respective donors were genotyped for CYP 1B1 (C432G) polymorphism by rt-PCR and the influence on outcome was analyzed. Median follow-up was 76 months. Results: 169 patients (44%) were genotyped as homozygous wild-type (wt) gene C/C, 157 (41%) as heterozygous genotype C/G and 56 (15%) as homozygous gene mutated G/G. Patients’ demographic, disease and treatment characteristics were equally distributed and showed no difference between the three subgroups (C/C, C/G and G/G) except that CYP 1B1 C/C was more common in females (52%) than in males (38%), p=.02. Five-year estimate for overall survival (OS) was 58% ±4 for the C/C group and 48% ±3 for the C/G- and G/G groups (p=.036). Surprisingly, this difference was primarily evident in males (p=.024), where the group with CYP 1B1 gene mutations did significantly worse: 58% ±6 vs. 42% ±4, whereas it was virtually absent in females. Transplant-related mortality (TRM) in male patients was higher for the group with mutated (mut) genes compared to the group with wt gene: One-year estimate for TRM 15% ±3 C/C vs. 20% ±3 C/G vs. 38% ±4 G/G (p=.048). Three-year estimate for relapse rate in male patients was 31% ±6 (wt) vs. 41% ±5 (mut), p=.097. Interestingly, male patients had more relapses than female patients if mut recipient SNP 41% ±5 vs 24% ±5 (p=.007) or mut donor SNP. Multivariate analysis (Cox regression) for OS in male patients revealed only two prognostic factors: mut donor SNP relative risk (RR) 1.4 (CI: 1.1–1.9), p=.007 and advanced disease RR 2.8 (CI: 1.9–4.1), p=.000. Conclusion: These results suggest that male patients with genetic polymorphism of CYP 1B1 have a lower OS due to a higher TRM and relapse rate after HSCT. Genotyping for CYP 1B1 (C432G) might help to identify patients with higher risk for allogeneic HSCT and may help explain gender differences in outcome. Prospective studies or confirmation in independent datasets are warranted. Disclosure: No conflict of interest disclosed.

44

Oncol Res Treat 2015;38(suppl 5):1–270

V163

Hepatic iron overload predicts for poor long-term overall survival in AML and MDS patients undergoing allogeneic stem cell transplantation Eckoldt J.1, von Bonin M.1, Sockel K.1, Middeke M.1, Stölzel F.1, Schetelig J.1, Ehninger G.1, Theurl I.2, Bornhäuser M.1, Platzbecker U.1, Wermke M.1 Uniklinik Dresden, Medizinische Klinik I, Dresden, Germany, 2Uniklinik Innsbruck, Department für Innere Medizin VI, Innsbruck, Austria 1

Background: Iron overload (IO) is a frequently observed condition in AML and MDS patients undergoing allogeneic stem cell transplantation (allo-SCT). Its definition and prognostic implications are a matter of constant debate. We have shown recently, that IO characterized by a liver iron content (LIC) ≥125 µmol/g (7 mg/g) as assessed by magnetic resonance imaging (MRI) correlated with a significantly increased early non-relapse mortality. However, this study as well as all other LIC-based studies published thus far, were characterized by a short follow up leaving the impact of IO on long-term outcomes such as overall survival (OS) and chronic GvHD (cGvHD) elusive. Patients and methods: Here we report the long-term outcome of our previously published cohort of 64 AML and 24 MDS patients undergoing allo-SCT. Pre-transplantation IO was measured by serum ferritin, transfusion burden or liver MRI. Results: After a median follow-up of 41 (range 4–66) months, IO defined by a LIC of ≥125 µmol/g was associated with a significantly shortened overall survival (Figure 1, p = 0.013). Elevated LIC was identified as an independent adverse risk factor for OS in a multivariate Cox regression model (HR 2.15, 95% CI 1.16–3.98, p = 0.015), whereas an increased serum ferritin above 2500 ng/ml or a transfusion burden of ≥20 red blood cell concentrates (RBC) did not predict for adverse prognosis. Hepatic IO was not associated with an increased likelihood of relapse (3 year cumulative incidence: IO: 36.6% vs. non-IO: 41.9%, p = 0.966) or cGvHD (3 year cumulative incidence: IO: 47.5% vs. Non-IO: 66.3%, p = 0.138). Infections after allo-SCT occurred at a similar frequency in IO (65.9%) and non-IO (68.2%) patients. Infections and GvHD were the most common causes of mortality each accounting for a third of all deaths without obvious differences between patients with and without an elevated LIC.

Fig. 1.

Conclusion: The increased early NRM observed in iron-overloaded AML and MDS patients undergoing allo-SCT translated into a reduced longterm overall survival. The causative relationship between IO and mortality warrants further investigations within prospective clinical trials.

Abstracts


CONTENTS AUTHOR INDEX

Disclosure: Julia Eckoldt: No conflict of interest disclosed. Martin Wermke: Expert Testimony: Novartis. V164

Post-transplant cyclophosphamide reduces haplo-identical transplant related mortality in non hodgkin lymphoma yielding similar results as with sibling donors Dietrich S.1,2, Finel H.2, Martinez C.2, Tischer J.2, Blaise D.2, Chevallier P.2, Castagna L.2, Milpied N.2, Bacigalupo A.2, Corradini P.2, Mohty M.2, Sanz M.2, Velardi V.2, Hausmann A.2, Montoto S.2, Hermine O.2, Schmitz N.2, Schouten H.2, Anna S.2, Robinson S.2, Dreger P.2 Medizinische Klinik V, Heidelberg, Germany, 2EBMT LWP, Paris, France

1

Background: Allogeneic stem cell transplantation (SCT) can provide long-term disease control for non hodgkin lymphoma (NHL), but its use depends on the availability of a matched donor. Haplo-identical relatives represent a reasonable alternative but data for haplo-transplants in NHL is sparse. Methods: Information of patients with mantle cell lymphoma (MCL), DLBCL, T-cell lymphoma (TCL) and follicular lymphoma (FL) who received an SCT from a sibling donor (SIB), 10/10 matched unrelated donor (MUD), haplo-identical donor (HAPLO) or cord blood (CORD) between 2007 and 2012 was downloaded from the EBMT database. Results: 2798 patients with NHL met the inclusion criteria. 2065 received a transplant from a SIB, 447 from a MUD, 167 from CORD (18 MCL, 36 DLBCL, 43 FL, 70 TCL) and 119 from a HAPLO donor (16 MCL, 30 DLBCL, 22 FL, 51 TCL). For 99 patients we received additional information on immunosuppression details after haplo-transplantation. 60 patients were treated with post-transplant cyclophosphamide (post-SCT cy) and 39 patients received other haplo-protocols. Patient characteristics were balanced except for higher median age in the post-SCT cy group. OS was significantly better for post-SCT cy treated patients than for alternative haplo-protocols (p = 0.0052, HR 2.3). More non relapse mortality (NRM) related deaths among alternative haplo-transplants accounted for the difference in OS (p = 0.032, HR 1.8), whereas relapse rates were similar. Observed trends for better OS of post-SCT cy treated patients held separately true for B- and T-cell lymphomas. Comparison of post-SCT cy haplo-transplants with alternative donor types demonstrated similar OS of SIB-, MUD- and post-SCT cy haplo-transplants, although haplo-transplants were performed in more advanced disease stages, patients had poorer performance status and were older. OS of CORD and alternative haplo-transplants were significantly worse than for SIB, MUD and post-SCT cy haplo-transplants (p < 0.001), which was attributable to lower NRM incidences for SIB, MUD and postSCT cy haplo-transplants (p < 0.001). Acute severe GvHD (grade 3–4) incidences were not different between donor groups, but post-SCT cy haplo-transplants had a trend towards lower chronic GvHD incidences. Conclusions: Haplo-identical donors and immunosuppression with postSCT cy is a valuable alternative to SIB or MUD transplants in NHL patients whereas CORD- and alternative haplo transplants are associated with increased mortality.

and intensified GvHD treatment changed the landscape of allogeneic transplantation. Here, we analyzed the influence of several parameters on the development of the cellular and humoral immunity of 136 patients consecutively transplanted in a single center. Methods: All adult patients (median age 52 years) transplanted over three years starting in September 2010 were analyzed retrospectively in respect of routinely measured immunoglobulin levels (turbidimetric method) and lymphocyte subset counts (FACS staining). Only in 9% of patients full conditioning regimen was used; nearly all patients received either anti-thymoglobulin (ATG) or alemtuzumab (25/136 matched related donors) leading to a relatively low rate of chronic GvHD (40%). The immune parameters achieved one year after stem cell transplantation were correlated with the conditioning regimen, diagnosis, relapse, age, sex and history of CMV infection, acute and chronic GvHD using Pearson correlation, t-test and regression analysis (SPSS). Results: A normal CD4/CD8 ratio at day +360 was only seen in 14% of available patients (3% elevated, 83% reduced) mostly caused by impaired CD4 cell count (90% < normal) while CD8 cell count was increased in 42%. The IgG level was normal in 51%, reduced in 31% and increased in 17% of tested patients. The CD8 cell count was significantly higher in patients after CMV infection (p < 0.001) and without alemtuzumab (p = 0.017) in the conditioning regimen; the IgG level correlated with history of CMV infection, CD4, NK (all p = 0.002) and CD8 (p = 0.025) cell count; age, sex, diagnosis, relapse, acute and chronic GvHD had no significant impact. Conclusions: Despite progress in recent years nearly all patients show a compromised immune system one year after allogeneic stem cell transplantation mainly characterized by a reduced CD4 cell count. In contrast, the CD8 cell count is often elevated probably reflecting response to viral infections. Interestingly, the immunoglobulin levels are highly variable ranging from antibody deficiency to hyper-immunoglobulinemia positively correlating with previous CMV infection. Surprisingly, GvHD had only a minor impact on the immune reconstitution of the investigated collective. Disclosure: No conflict of interest disclosed. V166

Regulatory B-cells can be identified early post-transplant and are raised in low grade acute Graft versus Host disease Chakupurakal G.1, Garcia-Marquez M.1, Shimabukuro-Vornhagen A.1, Schloesser H.2, Theurich S.3, Scheid C.1, Hallek M.1, Holtick U.1, von Bergwelt-Baildon M.1 Uniklinikum Koeln, Innere Medizin I, Koeln, Germany, 2Uniklinikum Koeln, Klinik für Allgemein-, Viszeral- und Tumorchirurgie, Koeln, Germany, 3Uniklinikum Koeln, Max-Planck-Institute for Metabolism Research, Koeln, Germany 1

Introduction: The renewal of the immune system after allogeneic stem cell transplantation is a complex process not fully understood. Recently the use of reduced intensity conditioning regimens, new anti-viral drugs

Allogeneic stem cell transplantation (alloSCT) is the curative therapeutic option for a variety of haematalogical malignancies. Graft versus Host disease (GvHD), with an incidence of around 40–60%, remains a major post transplant complication. The role of B-cells and especially the Interleukin-10 (IL-10) producing regulatory B-cells (B10-cells) in the pathophysiology of GvHD is not clearly understood to date. We studied B cells in allogeneic transplant recipients in the early post transplant phase. Samples, on 95 patients, transplanted at the University of Cologne and 10 healthy donors were collected prospectively after obtaining informed consent Patients were transplanted mostly for haematological malignancies (n = 94). Male: Female = 52:43. 86/95 90% received RIC regimens (90%). No patient received a B-cell depleting agent in the conditioning regimen or in the 6 weeks prior to transplant . 43 (46%) had no GvHD, and 10 (10%), 9 (9%),17 (18%)and 16 (17%) had grade 1–4 GvHD as per the modified Glucksberg criteria 6 respectively. Day 30 post transplant the total B-cell percentages in transplant recipients were significantly reduced in comparison to the controls (p=< 0.0001). Transitional B-cells were significantly more in transplant recipients (p = 0.004). Omission of data on the MAC patients did not affect the results. B10-cells could be demonstrated in 22 transplant recipients studied

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. V165

CMV infection plays a crucial role in the immune reconstitution after allogeneic stem cell transplantation Hartjen A.S.1, Krumbholz A.2, Thieme F.3, Bulduk M.1, Humpe A.1, Gramatzki M.1, Günther A.1 CAU / UKSH Kiel, Sektion für Stammzell- und Immuntherapie, II. Med. Klinik, Kiel, Germany, 2CAU / UKSH Kiel, Institut für Infektionsmedizin, Kiel, Germany, 3 UKSH, Institut für Klinische Chemie, Kiel, Germany 1

45


CONTENTS AUTHOR INDEX

(12 with acute GvHD). The percentages of B10-cells were significantly reduced in the transplant recipients on comparison with the control cohort (p < 0.0001). The mean percentage of B10-cells in patients with acute GvHD (1.94±1.2), though lower than control cohort 5.01±2.2, was significantly higher than those without GvHD 0.84±0.6 (p = 0.0003). This is the first study of B-cells and B-cell subsets suggesting that B-cell reconstitution commences immediately following engraftment. This is the first report demonstrating B10-cells in stem cell transplant recipients in the early post transplant (30 days) period and that they are enriched within the transitional cell compartment. Contrary to expectations, the percentage of B10-cells in patients with GvHD was significantly higher than those without GvHD (Fig 2C). The frequency of B10 cells were significantly higher in patients with lower grade GvHD suggesting a parallel to the observation on T-regs- where higher the frequency of T-regs the lower the GvHD grade and lower the non-relapse mortality (NRM) associated with an alloSCT. Disclosure: No conflict of interest disclosed.

Expertenseminar

Leichtketten-Amyloidose: Aktueller Stand der Diagnostik und Therapie V169

Light-chain amyloidosis: Current status of diagnostic and treatment Hegenbart U.1, Schönland S.1 Universitätsklinikum Heidelberg; V. Medizinische Klinik, Amyloidose-Zentrum, Heidelberg, Germany 1

Systemic amyloidoses are rare life-threatening disorders. Misfolded proteins which circulate in the blood are deposited in several organs (excluding the central nervous system) leading to organ dysfunction or failure. The prognosis is still poor: patients with advances cardiac involvement may die within 6–12 months of cardiac failure. There are various causes which must be differentiated. Amyloidoses are discriminated due to the causative pathologic protein (e.g. light chain, SAA or transthyretin) and in hereditary and non-hereditary forms. The amyloid light-chain (AL) amyloidosis is most common in Europe. In Germany about 800 patients will be diagnosed per year. A certain differentiation of the amyloid types is essential to avoid wrong treatment. First symptoms are non-specific; therefore the diagnosis is often (very much) delayed. Due to the availability of new diagnostic tools and treatments prognosis has improved; however the early diagnosis plays a crucial role to avoid organ failure or death of the patients. Goal of the chemotherapy is to destroy the clonal plasma cells. The missing replenishment of amyloidogenic light chains is able to stop the pathologic process of protein deposition which leads to improvement of organ function and quality of life in a large number of patients (within months to years). The multidisciplinary collaboration of several medical disciplines has an outstanding role to care for those severely ill patients. Disclosure: Ute Hegenbart: Financing of Scientific Research: Janssen, Celgene, Binding Site. Stefan Schönland: Financing of Scientific Research: Janssen, Celgene, Binding Site; Expert Testimony: Janssen, Celgene.

Posterdiskussion AML P170

Feasibility of semi-automated MRD analysis by flow cytometry in acute myeloid leukemia Köhnke T.1,2, Rechkemmer S.1, Bücklein V.L.1,2, Hiddemann W.1, Subklewe M.1,2 LMU München, Medizinische Klinik und Poliklinik III, München, Germany, Klinische Kooperationsgruppe Immuntherapie am Helmholtz Zentrum München, München, Germany 1 2

Introduction: The relevance of minimal residual disease (MRD) detection in AML has increased within the past decade and currently, clinical trials are evaluating the value of MRD-guided therapy. Analysis of this data is performed manually, by creating gates defining a leukemia-associated immunophenotype (LAIP) at primary diagnosis, which is then applied to follow-up samples. However, antigen expression has been shown to display some variability during therapy, which might reduce the sensitivity of this strategy. Methods: To test the feasibility of the recently proposed viSNE (visualization of t-distributed stochastic neighbor embedding) algorithm to detect aberrant populations, we analysed patient samples acquired within 3 months prior to hematological relapse, which were MRD-negative by traditional LAIP-gating. 400.000 events were acquired using an 8 color, 10 parameter panel on a Navios® flow cytometer (Beckman Coulter, Brea, CA, USA). For the bioinformatic analysis, the patient’s sample and a healthy donor sample measured on the same machine were combined digitally and viSNE clustering was applied to the resulting dataset. As previously suggested, MRD positivity was defined as the presence of a distinct cluster of >100 cells which consisted of >90% patient cells. Results: From our database, we identified 12 patients with comprehensive MRD flow assessment available within 3 months prior to hematological relapse who were in CR by cytomorphology at the time of MRD assessment. 9 of these patients were MRD-positive by traditional LAIP-gating (>0.1%). This first analysis therefor focused on the remaining 3 patients with MRD-levels at 0.01%, 0.03% and 0.07% at 21, 42 and 37 days prior to hematological relapse. Using viSNE, distinct clusters of 5058, 225 and 1043 events consisting of 95.5%, 92.6% and 93.8% patient cells were identified, consistent with MRD positivity as defined above. In all three cases, the cells identified by the viSNE algorithm showed the immunophenotype seen at relapse, while there were immunophenotypical shifts compared to primary diagnosis, possibly explaining false-negative MRD-assessment by traditional gating. Conclusion: viSNE clustering detected minor, aberrant populations in 3 patients with subsequent hematological relapse, which were not detected by traditional LAIP-gating strategy. viSNE might therefor aid in the analysis of high-dimensional flow cytometry data to detect small, aberrant populations. Further validation is ongoing. Disclosure: No conflict of interest disclosed. P171

LSD1 inhibition induces differentiation in murine models of AML Barth J.1, Scheder A.-M.2, Schulz-Fincke J.1,3, Schmitt M.3, Walter A.3, Lübbert M.4, Jung M.3, Serve H.2, Berg T.1,2 German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany, 2Goethe Universität Frankfurt, Medizinische Klinik II – Hämatologie/Onkologie, Frankfurt, Germany, 3University of Freiburg, Institute of Pharmaceutical Sciences, Freiburg, Germany, 4University of Freiburg, Department of Medicine I, Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany 1

The histone demethylating enzyme LSD1 represents a promising epigenetic target in the treatment of AML and inhibition of LSD1 has been shown

46

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

to facilitate a response of AML cells to all-trans retinoic acid (ATRA). In this project, we have now modeled the effect of pharmacological inhibitors of LSD1 in different murine leukemia models. Hematopoietic progenitor cells isolated from murine bone marrow were transformed by retroviral overexpression of a combination of Hoxa9 and Meis1 or MN1 turning them into cytokine-dependent progenitor cultures that elicit AML upon transplantation into mice. We have then used these cells to study the effect of different pharmacological inhibitors of LSD1. In detail, we treated the Hoxa9/Meis1- and MN1-transformed cells with three irreversible (tranylcypromine, AW69, AW84) and two reversible LSD1 inhibitors (MS120 (=Namoline), MS142) alone or in combination with ATRA. AW69 and AW84 reduced the short-term proliferation of MN1 cells by a maximum of 50% at day 3 of treatment, while the proliferation of Hoxa9/ Meis1 cells was not affected by any of the inhibitors. When analyzing the morphology of Hoxa9/Meis1 cells after treatment with AW69, AW84 or high-doses of tranylcypromine we observed signs of granulocytic differentiation. Flow cytometric analysis showed an increase of Mac1 and a reduction in CD117 (C-kit) after treatment with AW69 in Hoxa9/Meis1 and an increase in the expression of FcERI in MN1 cells. Colony formation of Hoxa9/Meis1 cells treated with AW69 for 96hrs was reduced by 70% compared to control cells. Treatment with AW69 and AW84 also enhanced the response of leukemia cells to ATRA by increasing the anti-proliferative effect and by reducing colony formation by 70% in MN1 and by 99% in Hoxa9/Meis1 cells in the combination. The two reversible LSD1 inhibitors MS120 and MS142 exhibited only minor effects on differentiation even when used at high concentrations. We conclude that pharmacological inhibition of LSD1 by tranylcypromine and its derivatives induces differentiation in Hoxa9/Meis1 cells and in MN1 cells. It is now planned to assess the effect of LSD1 inhibitors on leukemic stem cells and in vivo. This project will allow to select new LSD1 inhibitors for treatment and facilitate molecular studies investigating the role of LSD1 in the development of AML. Disclosure: No conflict of interest disclosed. P172

Dronabinol leads to disease control via release of differentiation blockage in an elderly patient with secondary acute leukemia Kampa-Schittenhelm K.1, Kanz L.1, Schittenhelm M.M.1 Universitätsklinikum, Tübingen, Germany

1

Dronabinol displays considerable growth-inhibiting antitumor efficacy in several tumor models and we have recently shown a direct proapoptotic effect in native leukemia patient blasts ex vivo. As we discuss separately, we have evidence that these effects are at least in part mediated via epigenetic hypomethylation (separate abstract provided). Here we report on a multimorbid elderly patient with secondary acute myelomonocytic leukemia treated with dronabinol for best supportive care considerations due to tumor kachexia and emesis. After initial cytoreduction with hydroxyurea no further specific cytoreductive therapy was administered and the patient was referred to palliative home care. Tantalizingly, while still taking dronabinol, the patient recovered and presented two months later with normal blood counts and absence of leukemia blasts. To confirm that clinically relevant doses are achievable in vivo, we set up a plasma inhibitory assay and cultured referenced Jurkat cells in serum of this patient. A strong proapoptotic effect of approx. 50% was noted after 72 hours of incubation. Further follow up of this patient revealed a steadily increasing monocyte count, suggesting leukemia relapse. However, cytomorphology confirmed adequately differentiating granulo- and monocytes and immunophenotyping revealed absence of CD34+ blasts (at diagnosis entire population reacting), while now being highly positive for CD13, CD14 and CD11c – indicating maturing cells. Interestingly in a leukemia mutation screen, an ASXL1 mutation was detected in this population, strengthening the

Abstracts

hypothesis that this maturing population stems from the former leukemia population. Notably, ASXL1 interacts with OGT, which is upregulated upon dronabinol (discussed in more detail in a separate protocol). In theory, upregulation of OGT may circumvent inactivating mutations of ASXL1 – and thus provide a stimulus for differentiation. Conclusions: Clinically active antileukemic doses of dronabinol can be achieved in vivo. Our findings provide a strong rationale for further exploration of dronabinol as an agent with remarkable antileukemic efficacy. Whether specific cohorts may in particular benefit from dronabinol (such as mut-ASXL1) needs to be addressed in future studies. Disclosure: No conflict of interest disclosed. P173

Response-adapted sequential azacitidine and induction chemotherapy in patients > 60 years old with newly diagnosed AML eligible for chemotherapy (RAS-AZIC): Results of the phase I of the DRKS00004519 study Jäkel N.1, Hubert K.1, Krahl R.1, Cross M.1, Niederwieser D.1, Al-Ali H.K.1 University Hospital of Leipzig, Department of Haematology/Oncology, Leipzig, Germany 1

Outcome in elderly patients (pts) with AML after intensive chemotherapy (IC) remains unsatisfactory. The hypomethylating agent azacitidine (AZA) prolongs OS in elderly pts even with >30% marrow blasts. But it is unlikely that a single agent could accomplish the goal of attaining rapid responses and translating them into long-term survival in all pts because of the heterogeneity of the disease. Thus, integrating epigenetic therapy and IC in well-designed trials might further optimize outcome in elderly patients. The feasibility of priming with AZA prior to IC was studied in the phase I of the DRKS00004519 (RAS-AZIC) study. Patients and methods: RAS-AZIC is a prospective, multicentric, phase I/ II trial evaluating the feasibility (phase I) and efficacy (phase II) of priming with AZA followed by a sequential, and response-adapted therapy with AZA or IC in eligible pts > 60 years with newly diagnosed AML. The safety and the dose of priming with AZA 75 mg/m2/day s.c. for 5 (level 1) or 7 days (level 2) followed by IC on day 17 (Mitoxantrone/cytarabine) was established through a 3+3 design. The level at which not more than one of 6 pts experienced a dose limiting toxicity (DLT) would be used in the phase II. DLT was defined as an event at least possibly related to trial therapy within the first 56 days as following: Grade ≥ 3 liver or renal toxicity, or delay in WBC regeneration (≥ 1×109/L) beyond day 45 after IC. Results: No DLT was documented in both dose level 1 and 2. The phase I part was completed after enrolling 9 pts. Median age was 71 years. Secondary/therapy-related AML, and unfavourable cytogenetics were 56%, and 33% respectively. Baseline median marrow blasts were 44%. AZA priming was well-tolerated and given as out-patient therapy in 67%. In 8/9 pts IC could be started as per protocol. Median WBC regeneration > 1 x109/L following IC occurred after 24 days. No death till day 90 ensued. CR/CRi on day 56 was 78%. After a median follow-up of 13 months, median survival was not reached. Conclusions: The results of priming with AZA for 7 days followed by IC on day 17 in elderly pts with newly diagnosed AML were encouraging in terms of safety and efficacy. The phase II part is now actively enrolling. Disclosure: Nadja Jäkel: No conflict of interest disclosed. Haifa Kathrin Al-Ali: Financing of Scientific Research: honoraria and research funding.

Oncol Res Treat 2015;38(suppl 5):1–270

47


CONTENTS AUTHOR INDEX

P174

Leukemic cells from patients with acute myeloid leukemia (AML) functionally inhibit healthy CD34+ hematopoietic stem and progenitor cells (HSPC) Jäger P.1, Cadeddu R.-P.1, Zilkens C.2, Fenk R.1, Germing U.1, Kobbe G.1, Haas R.1, Schroeder T.1, Geyh S.1 Klinik für Hämatologie, Onkologie und Klinische Immunologie, Universitätsklinikum, Düsseldorf, Germany, 2Orthopädische Klinik, Düsseldorf, Germany 1

Introduction: Hematopoietic insufficiency is the hallmark of AML with cytopenia-related complications representing major causes of death. Albeit recent experiment evidence pointed towards an important role of the bone marrow (BM) microenvironment, the underlying mechanisms mediating hematopoietic insufficiency are still insufficiently understood. Since the BM of patients with AML is dominated by leukemic blasts accompanied by a reduction of normal CD34+ HSPC, we reasoned that leukemic cells might directly suppress normal HSPC. Methods: To experimentally address this hypothesis we modelled the situation of BM infiltration in vitro by exposing healthy BM-derived CD34+ HSPC to supernatants derived from leukemic cells. Conditioned media (CM) were harvested from 3 AML cell lines (THP-1, HL-60, MV4–11) as well as from a total of 23 newly-diagnosed AML patients covering all relevant WHO subtypes after 3 days of cultivation. Healthy CD34+ HSPC were incubated for 3 days in the presence of leukemic or control media. Subsequently, proliferation, cell cycle behaviour and differentiation of these CD34+ HSPC was investigated using cell counting, dye staining with Ki-67 and Hoechst 33342 as well as semisolid clonogenic assays. Results: Exposure to conditioned media derived from AML cell lines and primary patients samples significantly inhibited proliferation as indicated by a profound reduction of viable healthy CD34+ HSPC. Complementary with this, we observed a clear shift of the cell cycle state of healthy CD34+ HSPC towards a resting phenotype when cultivated in AML-derived media with the majority of cells being in inactive G0 phase. Performing semisolid clonogenic assays demonstrated a strikingly lower colony-forming capacity of CD34+ HSPC following incubation with AML-derived supernatants. These inhibitory effects on healthy hematopoiesis were markedly related to the CD34+ leukemic cell population, but not to the MNC fraction as indicated by a comparison of paired MNC and CD34+ immunomagnetically enriched AML samples. PCR-screening of well-known negative regulators of hematopoiesis revealed a significant overexpression of TGF beta suggesting a potential role of this candidate molecule for suppression of healthy hematopoiesis by leukemic cells. Conclusion: Overall, these data indicate that leukemic cells mediate direct suppressive effects on important functions of healthy CD34+ HSPC thereby contributing to hematopoietic insufficiency in AML. Disclosure: No conflict of interest disclosed. P175

CD33-BiTE® antibody construct mediated lysis of AML cells is influenced by the choice of the pretherapeutic cytoreductive agent in vitro Brauneck F.1,2, Krupka C.1,2, Lichtenegger F.S.1,2, Kufer P.3, Kischel R.3, Zugmaier G.3, Köhnke T.1,2, Altmann T.1,2, Schneider S.1, Fiegl M.1, Spiekermann K.1,4, Hiddemann W.1,4, Subklewe M.1,2,4 Department of Internal Medicine III, Klinikum der Universität München, Munich, Germany, 2Clinical Co-operation Group Immunotherapy at the Helmholtz Institute, Munich, Germany, 3AMGEN Research (Munich) GmbH, Munich, Germany, 4German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany 1

Introduction: In our previous work, we demonstrated that the CD33BiTE® antibody construct AMG 330 is able to induce activation and proliferation of autologous T cells and effectively mediates lysis of primary AML cells.

48

Oncol Res Treat 2015;38(suppl 5):1–270

We hypothesized that in AML patients with high leukocyte counts a cytoreductive phase prior to AMG 330 therapy might be beneficial to increase the E:T ratio and to reduce cytokine mediated toxicity. Ideally, the cytoreductive drug does not impair T-cell function. Methods: We evaluated the effect of cytarabine (20µM), azacitidine (1µM, 5µM) and hydroxyurea (10µM, 100µM, 1000µM) on T-cell proliferation and function in close analogy to potential treatment algorithms for AML. Healthy donor T cells were pre-incubated with the cytoreductive drugs for 72 hours and subsequently co-cultured with HL60 cells with or without AMG 330. After 3 days, AMG 330 mediated lysis of AML cells and T-cell proliferation were assessed by flow cytometry. Results: Pretreatment of T cells with cytarabine completely abrogated T-cell function (lysis of HL60 cells: untreated (no tx): 98.2% vs 20µM: 7%) and significantly impaired T-cell proliferation (no tx: 57.4% vs 20µM: 4.3%, n = 3). These findings correlated to data using primary AML samples collected 3 and 6 days after discontinuation of cytarabine treatment in vivo. After a 3-day chemotherapy-free interval, we observed no relevant T-cell proliferation and lysis of AML cells upon addition of AMG 330 to the ex-vivo long-term culture system (lysis of AML cells on day 12: 30%; fold change T-cell expansion 0.9). After a 6-day treatment-free interval, high T-cell proliferation and cytotoxicity were observed (lysis of AML cells on day 12: 61%; fold change T-cell expansion: 3.1). Azacitidine treatment only marginally impaired T-cell function (lysis of HL60 cells: no tx: 100% vs 1µM: 94.9% vs 5µM: 86.8%; proliferation: no tx: 90.9% vs 1µM: 80% vs 5µM: 66.8%). Pretreatment with hydroxyurea had the least impact on T-cell performance. It did not impair T-cell function (lysis of HL60 cells: no tx: 99.9% vs 10µM: 96.5% vs 100µM: 97.7% vs 1000µM: 99.9%) and proliferation compared to untreated controls (no tx: 79.5% vs 100µM 74.6% vs 10µM 75.2% vs 1000µM: 81.9%, n = 3). Conclusion: As the BiTE® antibody construct technology essentially relies on T-cell function chemotherapeutic approaches need to be carefully evaluated. Our data support the use of hydroxyurea in AML patients that require cytoreduction prior to AMG 330 treatment. Disclosure: Marion Subklewe: received research funding from AMGEN Research (Munich) GmbH P176

The gain-of-function KIT D816V mutation associated with systemic mastocytosis (SM) is sensitive towards the FLT3 inhibitor crenolanib (Cb) and sensitizes cells towards cladribine (2-CdA) – a rationale to combine 2-CdA with Cb Kampa-Schittenhelm K.1, Frey J.1, Ramachandran A.2, Kanz L.1, Schittenhelm M.M.1 Universitätsklinikum, Tübingen, Germany, 2AROG Pharmaceuticals Inc., Dallas, United States 1

Activating D816 mutations of the class III receptor tyrosine kinase KIT are associated with the majority of patients with SM, making mutant-KIT an attractive target using tyrosine kinase inhibitors (TKI) – however, so far clinical efficacy of TKI fell short of the expectations. Cb is a novel potent FLT3 inhibitor with decent co-activity against the imatinib-resistant KIT D816 isoforms (Kampa-Schittenhelm DGHO 2014), however combination regimens might increase clinical efficacy. We have evidence that gain of the D816V enzymatic pocket type mutation within the tyrosine kinase domain 2 (TK2) sensitizes mast cells to 2-CdA – making it an attractive compound to combine with Cb. HMC1.1, HMC1.2 and the p815 mastocytosis cell lines were treated with Cb and/or 2-CdA. Cell cycle analyses and induction of apoptosis were performed flow cytometrically. Antiproliferative efficacy was measured using XTT-based assays. Tyrosinphosphorylation of KIT and downstream signaling was assessed by western immunoblots. For synergy analysis isobolograms were computed using Calcusyn software. HMC1.1 cells, harboring a regulatory KIT V560G mutation, treated with 2-CdA in a dose-dependent manner demonstrated a potent antiproliferative effect in the hundred nanomolar ranges – surprisingly, no significant proapoptotic effect was observed. We therefore evaluated time- and

Abstracts


CONTENTS AUTHOR INDEX

dose-dependent cell cycle effects and observed a dose-independent permanent G2/M phase arrest. Consequently, combination of 2-CdA with Cb (which accumulates cells in G1/G0 as most class III TKI) did not enhance proapoptotic effects. Interestingly, the sister cell line HMC1.2, harboring an additional D816V mutation displayed cell cycle independent high antiproliferative as well as proapoptotic sensitivities towards 2-CdA. High proapoptotic efficacy for 2-CdA was confirmed in murine p815 cells (harboring a D814Y mutation, corresponding to the D816 codon in humans). Combination of 2-CdA with Cb resulted in an additive proapoptotic effect, compared to either substance alone. It has been shown, that knockdown of p21 leads to resistance towards 2-CdA in colorectal tumor models. Tantalizingly, p21 is regulated via the PI3K/AKT pathway, which is directly activated via the KIT TK2 domain, providing a mechanistic model for our observation – in detail currently evaluated by our group. To summarize, we provide a rationale to combine 2-CdA and Cb in KIT D816 positive SM. Clinical evaluation is warranted. Disclosure: No conflict of interest disclosed. P177

The Adult Comorbidity Evaluation-27 (ACE-27) score is associated with survival in patients with acute myeloid leukemia Hitz F.1, Müller-Tidow C.1, Müller L.P.1, Wass M.1 Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany 1

Introduction: The role of comorbidities as an independent prognostic factor for patients with AML is ill defined. The ACE-27 comprises a tool to assess comorbidities. The aim of this study was to determine the impact of pretreatment comorbidities categorized by ACE-27 on the survival of patients with AML. Methods: In a single-center retrospective study the ACE-27 was obtained for 194 adult AML patients (median age 61y) treated with intensive chemotherapy between 1996 and 2012. Data on demographics, cytogenetics according to ELN and outcome were collected. Kaplan-Meier methods and Cox regression were used to assess survival. Results: ACE-27 was 0 (none) in 20% of patients, 1 (mild) in 25%, 2 (moderate) in 38%, and 3 (severe) in 16%. The most frequent comorbidities were cardiovascular disease (60%), prior malignancy (39%) and diabetes mellitus (16%). The median overall survival was 17 months. However, it was associated with ACE-27 risk groups with 96, 18, 14 and 8 months for patients with none, mild, moderate and severe comorbidities respectively (p=.006). In univariate analysis, cardiovascular disease and renal impairment were associated with inferior survival (26 vs 12 months, p=.005; 17 vs 7 months, p=.016). Interestingly complete remission (CR) rates differed versus risk groups: 80%, 79%, 63% and 43% for patients with none, mild, moderate and severe comorbidities, respectively (p=.002). Also early death rate differed between patients with none or mild vs moderate or severe comorbidities (6% vs 14%). Multivariate analysis showed that higher ECOG score (HR 2.9, p=.003), poor cytogenetics (HR 3.8, p=.007) and ACE-27 ≥ 2 (p=.002) had a significant impact on overall survival. Multivariate adjusted HR were 2.1, 2.6 and 4.5 for mild, moderate and severe comorbidities, respectively, compared with no comorbidities. Interestingly, higher age (≥ 60 years) had no significant impact in multivariate analysis when comorbidities based on ACE-27 were included. Conclusion: Comorbidities assessed by ACE-27 seem to have a significant impact on survival of patients with AML. Patients with severe comorbidity had a greater than 50% decrease in survival, independent of age, ECOG and cytogenetics. A pretreatment assessment of severity of comorbidities according to the ACE-27 may help to identify patients with poor outcome. It appears that age at least partially impacts survival as a marker for comorbidities. Disclosure: No conflict of interest disclosed.

Abstracts

P178

Resistance to sorafenib in acute myeloid leukemia with FLT3-ITD may be overcome by down-regulating the nuclear repressor Ski by inhibitors of histone deacytelases Jehn L.B.1, Frech M.1, Metzelder S.K.1, Teichler S.1, Stabla K.1, Neubauer A.1 Department of Hematology, Oncology and Immunology, Philipps University of Marburg and University Hospital of Giessen and Marburg, Marburg, Germany 1

Acute myeloid leukemia (AML) is genetically a heterogenous disease. Besides age, cytogenetic and molecular aberrations are the most important prognosticators. AML with monosomy 7 or deletion of 7q (-7/del7q) has a poor prognosis and is associated with an up-regulation of the nuclear oncogene SKI. AML with -7/del7q reveal an up-regulation of SKI expression as microRNA29a, encoded on 7q32, regulates SKI-expression (Teichler et al., Blood 2011). Ski protein represses all-trans retinoic acid (ATRA) signaling and myeloid differentiation in AML cells in vitro via interacting with N-CoR (nuclear receptor corepressor) and cooperating with histone deacetylases (HDAC). This inhibitory effect of wild-type Ski can be abrogated by the HDAC blocking agent valproic acid (VPA) (Ritter et al., Leukemia 2006). Activating internal tandem duplication (ITD) mutations in the fms-like tyrosine kinase 3 (FLT3) gene (FLT3-ITD) with normal cytogenetics are associated with poor outcome in AML (Thiede et al., Blood 2002). The therapeutic options for relapsed or refractory FLT3-ITD-positive AML are limited, particularly in case of prior allogenic stem cell transplantation (SCT) or poor performance status. Here, the use of the multi-targeted tyrosine kinase inhibitor sorafenib is an effective treatment option with high response rates (Metzelder et al., Blood 2009 and Leukemia 2012). However, the development of drug resistance to this targeted intervention remains an important clinical problem. Statistical analyses of patients (n = 14) with normal karyotype and positive for the FLT3-ITD mutation revealed a significantly shorter survival with a high Ski protein level compared with patients with a low Ski protein level. We further have investigated the influence of HDAC inhibitors (HDACi) on sorafenib-sensitive and -resistant FLT3-ITD-positive AML cells with differing levels of Ski expression in vitro. Cells were treated with increasing concentrations of different HDACi. Cell viability was measured using trypan blue staining and MTT assay. Western analysis was performed to investigate changes in Ski expression during HDACi treatment. We found that high expression of Ski in sorafenib-resistant FLT3-ITD-positive cells was predictive of better responses to treatment with HDACi in vitro. In conclusion, the combined use of HDACi and multi-targeted tyrosine kinase inhibitors like sorafenib in relapsed or refractory FLT3-ITD-positive AML with high Ski expression deserves further consideration. Disclosure: No conflict of interest disclosed. P179

Prospective functional and patient-reported outcomes study of AML patients aged >60 years receiving induction chemotherapy (INCIDER trial): first feasibility analysis Maurer H.1, Müller M.J.1, Ihorst G.2, Bogatyreva L.1, Lübbert M.1 Universitätsklinikum Freiburg, Klinik für Innere Medizin I, Freiburg, Germany, Albert-Ludwig-Universität Freiburg, Department für Medizinische Biometrie und Medizinische Informatik, Freiburg, Germany 1 2

It is increasingly accepted that not only disease but also host-specific factors contribute to the treatment tolerance and outcome in elderly AML/ MDS patients (pts). We could previously show that patient-reported outcome parameters can predict overall survival of AML/MDS patients >60 year unfit for standard chemotherapy and therefore receiving non-intensive treatment or sole best supportive care (Deschler et al. 2013). Only very few studies have successfully shown that assessments also in AML pts fit for induction chemotherapy can yield prognostic parameters, e.g. Klepin et al. (Blood 2013). The INCIDER prospective study aims at defining functional and patient-reported outcomes parameters that might predict overall survival in this patient group.

Oncol Res Treat 2015;38(suppl 5):1–270

49


CONTENTS AUTHOR INDEX

Methods: Since 5/2012, all AML pts >60 years receiving induction chemotherapy at our center were screened for a pre-treatment assessment and up to 3 subsequent assessments. The following physician-assessed instruments were applied: G8 (geriatric symptoms), ECOG performance status, Barthel-index (Activities of daily living), HCT-CI (comorbidity burden). Patient-reported outcomes instruments: EORTC-C30 (quality of life), HADS (anxiety, depression), RS-11 (psychological resilience). Tests were applied by a team of 2 physicians and a study nurse. Results: 34 consecutive pts met the eligibility criteria for the study, of whom 25/34 (74%) were successfully assessed prior to induction (timepoint T0). Another 9/34 pts were not enrolled, reasons were as follows: 3 declined participation due to cognitive overload, 3 had rapidly progressive disease and evolving cognitive disability, 3 were not approached. Feasibility of subsequent assessments: at timepoint T1 (at 3 months) 15 of the 19 pts still alive were assessed; timepoint T2 (at 6 months): 12/14; timepoint T3 (at 12 months): 5/9. Thus the main drop-out reason was death. Conclusions: This feasibility analysis demonstrates that the assessment tests chosen were well accepted by most of the pts, and were feasible despite treatment initiations also on weekends. Also pts with rapidly progressive disease and difficult clinical conditions could be included by the team. Thus in this single-center pilot trial, patient compliance and logistics allowed for successful assessment of the majority of eligible pts. The study will be continued with particular attention to further improvement of patient coverage and timeliness of pre-therapy assessments, and expansion to more centers. Disclosure: No conflict of interest disclosed. P180

Successful treatment of acute promyelocytic leukemia in pregnancy with single agent ATRA Nellessen C.1, Mayer K.M.1, Merz W.M.2, Flöck A.2, Brossart P.1, Janzen V.1 Medizinische Klinik III, Hämatologie-Onkologie, Uniklinik, Bonn, Germany, Universitätsfrauenklinik, Abteilung für Geburtshilfe und Pränatalmedizin, Uniklinik, Bonn, Germany 1 2

Introduction: The diagnosis of acute promyelocytic leukemia (APL) in pregnancy is an uncommon, life-threatening emergency. Choice of treatment and management of complications is challenging, especially as published data are rare. Case: A 41-year old woman was referred to our emergency department at 25 weeks of gestation with spontaneous hematoma and pancytopenia. Diagnosis of APL was made by bone marrow cytology and confirmed by PCR. A delivery at that time was judged to be of unacceptable risk to the mother due to the presence of severe, APL-associated coagulopathy. Therefore, a joint decision was made to immediately start induction therapy with all-trans retinoic acid (ATRA) supplemented by dexamethasone to prevent ATRA differentiation syndrome. Induction treatment was continued with ATRA alone without addition of anthracyclines with the intention to minimize the risk of toxicity to the fetus. During the entire induction cycle no signs of differentiation syndrome were observed. Transfusion with red blood cells and platelets as well as substitution with clotting factors within the first few days was necessary and sufficient to prevent major bleeding incidents. At day 32 after initiation of therapy the blood cell counts had normalized, and the patient was discharged from hospital with continuation of ATRA therapy. Fetal wellbeing was assessed regularly by ultrasound and Doppler examination. Delivery was scheduled for 34 weeks of gestation and an apparently healthy female baby was delivered. Remission control of peripheral blood and bone marrow after delivery (day 72 of ATRA treatment) showed complete cytomorphologic and molecular remission. We immediately proceeded with the first consolidation course of ATRA and arsenic trioxide. Currently (May 2015), the patient is in her third consolidation course, remaining in complete remission. Conclusion: This case illustrates that treatment with ATRA alone in the second half of pregnancy was safe for both mother and fetus, achieving complete remission of APL and delivery of a healthy newborn. Close sur-

50

Oncol Res Treat 2015;38(suppl 5):1–270

veillance of both, mother and fetus, and management of disease related complications, and interdisciplinary teamwork with obstetricians was mandatory for successful outcome. Disclosure: No conflict of interest disclosed. P181

Metabolic reprogramming of acute myeloid leukemia blasts by bone marrow stromal cells Braun M.1, Jitschin R.1, Mackensen A.1, Mougiakakos D.1 Universitätsklinikum Erlangen; Hämatologie und Onkologie, Erlangen, Germany 1

Acute myeloid leukemia (AML) represents the most common form of acute leukemia in adults. Despite the enormous efforts during the last decades treatment resistance is still observed at a high rate. Previous studies have shown that bone marrow stroma promotes an increased resilience of AML blasts towards chemotherapeutics. Furthermore, current data suggest that alterations of the malignant cells’ metabolism could represent a strong determinant for the disease’s (including AML) course and/or treatment resistance. In fact, a deregulated metabolism could lead to a reduced sensitivity towards therapy and it remains to be elucidated whether this is a mechanism contributing to the blast-protective effects elicited by the bone marrow stroma. Here, we sought out to characterize the impact of stroma cells on the AML blasts’ metabolism. The human bone marrow stromal cell line HS-5 was utilized for establishing the in vitro niche model. We compared in our assays AML cell lines as well as primary blasts cultured on a HS-5 monolayer or alone. In line with previous observations we could detect an increased proportion of AML cells in the S-phase of the cell cycle upon co-culture with HS-5. When evaluating the cells’ metabolism we observed a shift towards glycolysis despite presence of oxygen, i.e. aerobic glycolysis or the “Warburg”-effect. Basal glycolysis as well as maximal glycolytic capacity upon blocking ATP production in the mitochondrial respiratory chain was increased. Respiration was not significantly affected. However, mitochondrial biogenesis appeared reduced. Increased glycolysis was accompanied by an increased uptake of fluorescently labeled glucose as well as an increased expression of glucose transporters. The expression of several glycolytic molecules found to be increased upon HS-5 co-culture. Noticeably, cell-to-cell contact was not a pre-requisite for the metabolic shift. Our data was further corroborated by direct observations from AML patients: AML blasts isolated from the bone marrow exhibited an upregulated glycolysis as compared to their counterparts from the periphery collected at the same time point. Taken together, our data indicates a stromal cell-mediated metabolic shift in AML blasts towards aerobic glycolysis. This metabolic phenotype is linked to an unfavorable prognosis and increased chemo resistance. The underlying molecular pathways remain to be elucidated and could represent a promising target for future interventions. Disclosure: No conflict of interest disclosed. P182

Blastic plasmacytoid dendritic cell neoplasm: A very rare, unfamiliar and scary disease Schmidt A.1, Neuberger C.2, Schmid M.1, Lienhard R.3 Stadtspital Triemli, Internal Medicine, Zürich, Switzerland, 2Stadtspital Triemli, Pathology, Zürich, Switzerland, 3Spital Affoltern am Albis, Internal Medicine, Affoltern am Albis, Switzerland 1

Introduction: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a very rare haematological disease currently categorized as an entity of “acute myeloid leukaemia and related precursor neoplasm” in the WHO Classification of 2008.This malignancy is characterized by CD4+/CD56+/ CD123+ immunophenotype, aggressive course and poor prognosis. BPDCN often initially presents with solitary or multiple heterogenic skin lesions, accompanied by regional lymph node involvement. In advanced

Abstracts


CONTENTS AUTHOR INDEX

stages of disease, bone marrow infiltration and leukemic transformation occurs regularly. BPDCN is usually diagnosed by immunohistological staining. Therapeutic approaches are heterogeneous and despite high response rate to first line therapy, early relapse with disease progression and dismal prognosis is the rule. Methods: Based on an own case of an 71 year old man with at the beginning only cutaneous manifestation of BPDCN, we made an extensive literature research in PubMed for articles in several languages relating to small series and case reports of BPDCN. We reviewed 70 articles from 1997 to now considering initial clinical manifestation, chosen therapeutic regimen, evolution and survival. Special attention was laid to the time of starting therapy and use of novel substances like bortezomib. Results: Reviewing available literature (containing about 400 patients) confirmed BPDCN as a very rare disease of any age with heterogeneous clinical and histopathological presentation, often leading to misdiagnosis. Currently the only curative approach, showing long-term remission in selected cases, is allogeneic stem cell transplantation. Most patients were treated with ALL- or AML-like chemotherapy-regimen, associated with high response but early relapse rate, with a median survival of only approximately 12 months. Overall, evidence is scarce and newer drugs barely used. In our case, delaying start of therapy until needed and addition of Bortezomib to a conventional R-CHOP-regimen didn’t improve prognosis. Conclusion: BPDCN is a very rare disease with commonly inhomogeneous cutaneous manifestation that typically occurs in elderly patients. Currently no standardised treatment exists and early relapse with fulminant deterioration affects majority of patients. Further investigations and registries are needed to better understand pathophysiological processes of BPDCN and develop diagnostic and therapeutic guidelines to reduce misdiagnosis and improve long-term remission rate. Disclosure: No conflict of interest disclosed. P183

Acute promyelocytic leukemia resulting from a cryptic insertion responds well to ATRA plus ATO therapy: a case report Hecht A.1, Nolte F.1, Fabarius A.1, Haferlach C.2, Hofmann W.-K.1, Lengfelder E.1 Universitätsklinikum Mannheim, Hämatologie und Internistische Onkologie, Mannheim, Germany, 2MLL Münchner Leukämie Labor, München, Germany 1

Introduction: Normally, diagnosis of acute promyelocytic leukemia (APL) is based on the pathognomonic finding of the characteristic translocation t(15;17) resulting in the PML-RARA fusion gene. Submicroscopic cryptic insertions are rare abnormalities that can be found in approximately 3.6% of APL cases only (Grimwade et al., Blood 2000). Therefore, results on treatment response and outcome are solely based on case reports. Here we present a case of APL due to a cryptic insertion with an unusually favorable initial presentation and good response to ATRA plus ATO. Case report: In August 2014 an inquiry reached our international office concerning the treatment of a Ukrainian patient with APL. One week later a 27 year old woman presented with a history of prolonged fatigue. Diagnosis of APL had been based on a bone marrow (BM) smear one week before administration to our hospital. Though the patient had not undergone any kind of treatment, she did not present with any symptoms of bleeding or coagulation dysregulation. Laboratory results showed leukocyte count of 2,400/µl with no peripheral promyelocytes, hemoglobin level of 12.3 g/ dl and platelet count of 206,000/µl. Mild hypofibrinogenemia and subtly elevated levels of d-dimers were the only hints at coagulopathy. The initial BM smear showed a total infiltration by promyelocytic blasts with multiple auer rods. On the day of administration we performed another BM biopsy to confirm the results. Cytomorphology showed a 50% proportion of abnormal promyelocytes. Molecular genetic analysis proved the presence of the PML-RARA fusion transcript (long isoform). However, cytogenetic analysis showed a normal female karyotype. At last, FISH analysis re-

Abstracts

vealed a submicroscopic insertion of the RARA locus on chromosome 17 into the PML locus on chromosome 15 (ins(15;17)(q24;q21q21). The patient started treatment with ATRA and ATO according to the APL0406 protocol and achieved complete remission within the first 28 days. There were no APL or treatment related complications and she is now undergoing her last cycle of consolidation therapy. Conclusions: To our best knowledge this is the first report on response to the ATRA plus ATO combination therapy in an APL patient bearing a cryptic insertion. Interestingly, this patient also presented with an unusually benign course of APL. This adds knowledge to the few cases reported with this abnormality. Disclosure: No conflict of interest disclosed. P184

Dronabinol reveals antileukemic efficacy via epigenetic upregulation of O-linked β-N-acetylglucosamine transferase (OGT) resulting in release of differentiation blockage and sensitizing cells towards induction of apoptosis Kampa-Schittenhelm K.1, Salitzky O.1, Akmut F.1, Bonin M.2, Kanz L.1, Schittenhelm M.M.1 Universitätsklinikum, Tübingen, Germany, 2Universität Tübingen, Medizinische Genetik, Microarray Facility, Tübingen, Germany 1

Dronabinol, the natural (−)-Δ9-Tetrahydrocannabinol, has growth-inhibiting antitumor efficacy – including acute leukemia. We now reveal a novel mechanism-of-action via epigenetic modulation of OGT, an enzyme linked to genes involved in leukemogenesis such as AKT, MLL5, TET2 or ASXL1, releasing leukemia blasts from differentiation blockage in vivo and sensitizing cells towards induction of apoptosis. gDNA methylation gene arrays using Jurkat leukemia cells revealed global modulation of methylation patterns upon dronabinol treatment. OGT was identified as the highest altered gene (–42%, pval 3,68E-38) – correlating with an increase of OGT protein expression in Western immunoblots. Consistently, hypomethylation of the transcription start site of OGT and induction of OGT protein expression upon dronabinol were confirmed in an independent array using native patient samples. To study the underlying mechanisms, Jurkat cells were pretreated with CB1 (LY320135) and/ or CB2 (JTE-907) antagonists, and exposed to dronabinol. Inhibition of either receptor reduced induction of apoptosis – and was most profound when inhibiting both receptors simultaneously. Importantly, upregulation of OGT protein expression upon dronabinol was suppressed by inhibition of either receptor, arguing for a receptor-mediated epigenetic effect of dronabinol. Retroviral knockdown of OGT in Jurkat and native leukemia blasts rendered cells less susceptible towards induction of apoptosis. We had the chance to follow a patient with secondary acute myeloid leukemia who was supportively treated with dronabinol for tumor kachexia and emesis. Direct disease control linking to dronabinol was noted (case report provided as separate abstract). Tantalizingly, besides induction of apoptosis upon dronabinol, the leukemic clone was maturing – overriding the differentiation blockage. Mimicking this observation, we treated cells of this and other patients with dronabinol ex vivo and revealed loss of CD34 and upregulation of CD11c by flow cytometry – again a sign for maturation. Interestingly this differentiation was abrogated by lentiviral OGT-interference arguing for a role of OGT in overriding the differentiation blockage in acute leukemia. Conclusions: Our findings provide a strong rationale for further exploring dronabinol as an agent with remarkable antileukemic efficacy achievable in vivo. In specific, overriding the differentiation blockage in leukemia cells may open up alternative therapeutic approaches. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

51


CONTENTS AUTHOR INDEX

P185

Expression of inhibitory (i)ASPP (Apoptosis Stimulating Protein of p53) in acute myeloid leukemia (AML) Schittenhelm M.M.1, Bajrami Saipi M.1, Kanz L.1, Kampa-Schittenhelm K.1 Universitätsklinikum, Tübingen, Germany

1

Inactivation of the p53 pathway is a frequent event in human cancers promoting tumorigenesis and resistance to chemotherapy. Inactivating p53 mutations are uncommon in non-complex karyotype leukemias suggesting that the p53-pathway must be inactivated by other mechanisms. ASPP proteins are a family of p53-binding proteins that consists of three members: the pro-apoptotic ASPP1 and ASPP2 proteins and the apoptosis-inhibiting iASPP. All play a crucial role in the regulation of p53-induced apoptosis. We have shown that ASPP2, but not ASPP1, is an independent haploinsufficient tumor suppressor in vivo and is upregulated upon cellular stress. Further, ASPP2 is significantly attenuated in AML – and this uniquely links to the patient cohort failing induction chemotherapy. We now study changes of ASPP2 expression and its counterplayer iASPP during chemotherapy to evaluate whether ASPPs may provide a useful tool to predict and monitor therapy response. ASPP2 as well as iASPP mRNA levels were determined using standard qRT-PCR. Protein expression was measured using flow cytometry and immunoblot assays. Basal levels in bone marrow samples as well as in peripherel blood samples of so far 25 leukemia patients were normalized to a control cohort comprising 16 healthy bone marrow donor samples. Blood samples were obtained before and during initial therapy at defined timepoints (12, 24, 48 and 72 hours) and expression levels of ASPP2 and iASPP were measured and correlated with blast count and initial therapy response. We confirmed lower basal ASPP2 mRNA as well as protein levels in AML samples compared to the donor controls. During induction therapy, a proportion of patients revealed adequate upregulation of ASPP2 in vivo. Vice versa, we define a subgroup of patients that fail to upregulate ASPP2 during therapy. iASPP levels were generally higher in the patient compared to the donor cohort and tended to increase at the beginning of therapy. Interestingly, highest iASPP levels were found in patients expressing low ASPP2 levels – and these findings again correlated with poor clinical outcome. We believe, that different members of the ASPP family play a distinct role in acute leukemia and that complex interactions between the different family members are in part responsible and predictive for therapy outcome. Further studies are necessary to shed light into the role of iASPP in leukemia and to understand the complex interactions within the ASPP family. Disclosure: No conflict of interest disclosed. P186

Accelerated senescence of MSC from patients with high risk ALDH-numerous AML Horn P.1, Hoang V.T.1, Hoffmann I.1, Benes V.2, Wuchter P.1, Ho A.D.1 Universitätsklinikum Heidelberg, Innere Medizin V, Heidelberg, Germany, European Molecular Biology Laboratory, Genomics Core Facility, Heidelberg, Germany 1 2

The microenvironment in the bone marrow (BM) has significant effects on maintenance, regulation, and safeguarding of hematopoietic stem cells (HSC), both in health and development of leukemia. We have compared mesenchymal stromal cells (MSC) from patients with acute myeloid leukemia (AML) vs. healthy donors as surrogate niche models to analyze the environment of the leukemic stem cell niche. We compared gene expression profiles of AML samples derived from patients based on the expression levels of aldehyde dehydrogenase (ALDH) activity. MSC were isolated from human BM samples of healthy donors and AML patients with newly diagnosed AML with low frequencies of ALDH

52

Oncol Res Treat 2015;38(suppl 5):1–270

positive blasts (below median value of 1.92% for healthy bone marrow; ALDH-low) or high frequencies of ALDH positive cells (above 1.92%; ALDH-numerous). We have shown that the latter group of patients is associated with high-risk for relapse and poor clinical outcome. Cells were expanded for initial characterization, analysis of CFU-F frequency and long-term culture. RNA of early and senescent passages after long-term culture was used for gene expression profiling. The majority of AML-MSC displayed a senescent phenotype and shape, decreased CFU-F frequency, and a significantly decreased proliferation capacity, specifically those from ALDH-numerous AML. Comparison of genome profiles of MSC from patients with ALDH-numerous vs. ALDH-low AML showed differential expression of 1031 genes, 369 up- and 662 down-regulated. Of these over 62% could be directly related to senescence upon long-term cultures of MSC from healthy subjects, connected with GO-categories of various malignant disorders, and translation and gene Expression, 203 gene products being localized in the nucleus. Comparison of gene expression profiles of early and senescent cell passages of healthy MSC vs. early passages of AML-MSC showed a specific clustering of MSC derived from patients with ALDH-numerous AML with senescent passages of healthy donors. Profiles of MSC derived from patients with ALDH-low AML were similar to early passages of healthy donors. These data provide evidence that the microenvironment is involved in the malignant process of AML. The poor prognosis of ALDH-numerous AML is reflected by an accelerated senescence status and a characteristic gene expression profile of MSC derived from such patients. Experiments are ongoing to identify the roles of specific genes for the accelerated aging process in high risk AML. Disclosure: Patrick Horn: No conflict of interest disclosed. Anthony D. Ho: Advisory Role: Genzyme / Sanofi-Aventis; Financing of Scientific Research: Genzyme / Sanofi-Aventis. P187

Therapy related acute promyelocytic leukaemia – presentation of two cases Hoffmann F.1, Graul K.1, Heyn S.1, Franke G.-N.1, Schwind S.1, Jentzsch M.1, Pönisch W.1, Al-Ali H.K.1, Wang S.Y.1, Niederwieser D.1, Vucinic V.1 Leipzig University, Hämatologie und Internistische Onkologie, Leipzig, Germany 1

Introduction: Therapy related acute myeloid leukaemias (t-AML) account for about 10–20% of patients with AML and mostly occurs 5–10 years after treatment with alkylating agents and 2–3 years after treatment with topoisomerase II inhibitors. Here we present two patients with therapy related acute promyelocytic leukaemia (t-APL). Patients: Two female patients aged 49 and 58 years developed a t-AML after breast cancer treatment with 5-Fluorouracil (5-FU), etoposide and cyclophosphatide followed by two courses of docetaxel and sigma carcinoma treated with 5-FU, leucovorine and oxaliplatine, respectively. Patients presented with clinical symptoms of respiratory or urinary infection in April 2014 and January 2015, respectively. The blood counts in both cases showed a pancytopenia with platelet counts (PLT) above 100 GPT/l, i.e. low risk according to Sanz score. The bone marrow smear showed an infiltration with 40% and 45% blasts, respectively. In the fluorescent in situ hybridization (FISH) the positivity for t(15;17) was shown and confirmed by real time polymerase chain reaction (rt-PCR) method showing the PMLRARA fusion in both cases. The APL developed after 3 and after 5 years from diagnosis of the respective primary tumor. The initial treatment of t-APL in both patients consisted of a combination of all-trans-retinoic acid (ATRA) and an induction chemotherapy with intermediate dose cytarabine on days 1, 3, 5 and 7 and idarubicine on days 1, 2 and 3. Both patients achieved a complete remission (CR). The rt-PCR showed a 3 fold log reduction. In both patients a consolidation chemotherapy, identical to induction, was applied. The first patient showed a prolonged hematological regeneration but hematological CR was confirmed. The rt-PCR analysis showed no sign of disease. The patient is still in complete haematolog-

Abstracts


CONTENTS AUTHOR INDEX

ical and molecular remission, 1 year after diagnosis. The second patient is at the moment in neutropenic phase following the consolidation therapy. Discussion: The t-APL is more common in female patients, and the most common cases are patients with previously treated breast cancer. The t-APL after colonic carcinoma has been described in sporadic cases. In the published cases the t-APL developed shortly after diagnosis and treatment of the primary neoplasia (<3 years). t-AML has an equaly good prognosis as de novo APL, and until today both of the reported patients are in complete hematological remission. Disclosure: No conflict of interest disclosed.

Posterdiskussion Der spezielle Fall P188

Ruxolitinib as a treatment option for steroid-refractory severe graft-versus-host disease – case report Paul S.1, Wass M.2, Müller-Tidow C.2, Müller L.P.2 Universitätsklinikum Halle, Klinik für Innere Medizin IV, Halle, Germany, Universitätsklinikum Halle, Klinik für Innere Medizin IV-Hämatologie/ Onkologie, Halle, Germany 1 2

Introduction: If refractory to steroids, acute as well as chronic GvHD represent a life-threatening condition after allogeneic stem cell transplantation (SCT) for which no treatment standards exist. New data suggest that immunomodulatory drugs like ruxolitinib may be of therapeutic use in this situation. Case: We report on a 51-year-old male diagnosed with acute myeloid leukaemia with intermediate cytogenetic risk. After standard induction and consolidation therapy complete remission was achieved. In July 2014 myeloablative allogeneic SCT from an HLA-matched related donor was performed. Immunosuppressive therapy comprised cyclosporine and longcourse methotrexate without ATG. Upon rapid engraftment the patient developed severe, histologically proven acute GvHD of the skin (stage 4, IV°) on day +30. Therapy with prednisolone (2 mg/kgBW) was initiated without any response seen after 7 days. Therapy was escalated with cyclosporine, mycophenolate mofetil and ATG. Signs of GvHD improved completely but the patient developed pancytopenia, systemic EBV reactivation and septic shock. At day +100 severe GvHD of the skin relapsed. A second course ATG with prednisolone was applied followed by treatment with sirolimus, haploidentical mesenchymal stem cells and PUVA. Again treatment was accompanied by severe neutropenic septicaemia and EBV reactivation. After a short response a progression of cutaneous GvHD ensued. Based on a recent publication (Spoerl et al.: BLOOD 2014;123:3832) we started treatment with ruxolitinib at a dose of 10mg bid. Given the stable blood counts and lack of infections the dose was increased to 25mg bid. Treatment with prednisolone was tapered and on day +180 we observed a clear response with residual signs of moderate overlap GvHD. Conclusions: Steroid refractory GvHD represents a severe complication after allogeneic SCT for which no effective standard treatment is established and which therefore results in high mortality and morbidity. Our reported observation supports the further exploration of ruxolitinib as an effective treatment with low toxicity profile in such patients. Disclosure: No conflict of interest disclosed.

P189

Spectrum of hemophagocytic lymphohistiocytosis (HLH): three cases of acquired HLH with three different triggers Jung B.1, Hartmann U.2, Jaeschke B.1, Becker S.1, Märker-Hermann E.2, Frickhofen N.1 HSK, Dr. Horst Schmidt Klinik, Helios Kliniken Gruppe, Innere Medizin III (Onkologie, Hämatologie, Palliativmedizin), Wiesbaden, Germany, 2HSK, Dr. Horst Schmidt Klinik, Helios Kliniken Gruppe, Innere Medizin IV (Rheumatologie, Klinische Immunologie, Nephrologie), Wiesbaden, Germany 1

Introduction: Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening syndrome of excessive immune activation.The primary form manifests in children with genetic abnormalities of immune effectors. Secondary forms occur in the setting of conditions such as infection, malignancy, autoimmune disease or drugs. Methods: Case reports of 3 patients with acquired HLH, treated 2013– 2015. Diagnostic criteria were based on the HLH-2004 trial (Blood 118:4041;2011). Results: Patient 1: A 20yo female presented 10 days after tonsillectomy for tonsillitis with fever, erythema nodosum and hepatosplenomegaly. EBV-PCR suggested EBV infection as trigger of HLH.Treatment with dexamethasone 10 mg/m2 and 2 doses of etoposide 75 mg/m2 (dose reduced for impaired liver function) for 14 days overall led to complete resolution of signs and symptoms of HLH. Patient 2: A 61yo female with diffuse large B-cell lymphoma achieved PR after chemotherapy with cyclophosphamide, doxorubicin, liposomal vincristine, prednisone and rituximab. She experienced early progression and was treated with 2 courses of dexamethasone, high dose cytarabin and cisplatin (R-DHAP). After the second course she developed signs of HLH. It improved during treatment with dexamethasone 10 mg/m2 and 2 doses of etoposide 150 mg/m2, but the lymphoma relapsed. After a course of R-DHAX (cisplatin replaced by oxaliplatin for decreased kidney function) HLH relapsed again, now with severe neurological symptoms. Despite immediate initiation of dexamethasone / cyclosporine A , HLH progressed and the patient died. Patient 3: A 42yo female with a 15 months history of rheumatoid arthritis had been treated with methotrexate and adalimumab. 55 days after initiating adalimumab she presented with fever, aphasia, mental confusion, hepatosplenomegaly and rash. A diagnosis of HLH was made and she was started on dexamethasone/etoposide. Due to persistent fever etoposide was replaced by the IL1 receptor antagonist anakinra. Coincident with the start of anakinra and with PCR evidence of herpes and EBV viremia, HLH dramatically progressed with an increase of ferritin to 304.000 ng/ ml. Anakinra was replaced by cyclosporine A and high dose immunoglobulin and aciclovir was initiated. The patient recovered completely. Conclusions: These cases illustrate infection, lymphoma chemotherapy and adalimumab as triggers of HLH. Treatment of HLH aims to suppress the exaggerated immune response using corticosteroids, etoposide and cyclosporine A. Disclosure: No conflict of interest disclosed. P190

Imitation of bone metastasis by lymphangiomatosis in a patient with colon cancer Fund N.1, Nuber J.1, Schwella N.1 Klinikum, Ludwigsburg, Germany

1

Introduction: Lymphangiomatosis is a rare type of benign disease of the lymphatic system. In principle it can occur in any lymphatic tissue, but seems to prevalently appear in the region of thorax and neck. Even skeletal involvement might be possible. The disease mainly presents in childhood, but there are also cases described in the literature with occurrence in adults. Case report: A 38-year old male patient was admitted to the hospital because of anemia, significant weight loss and right abdominal pain. During

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

53


CONTENTS AUTHOR INDEX

a colonoscopy a malignant tumor of the colon ascendens was diagnosed. Histological findings revealed an adenocarcinoma. The following computer tomography of the thorax/abdomen/pelvis showed multiples osteolytic lesions in the cervical spine and in the pelvis. However, the skeletal scintigraphy remained without pathological findings. In addition a large cystic lesion of 10 × 14 cm was detected within the left upper mediastinum. The differential diagnosis of the mediastinal mass suspected a dermoid cyst or a pleural effusion. However, because of the multiple osteolytic lesions, we assumed a palliative disease related to a metastasized cancer of the colon. Due to the stenosing malignant process, the patient underwent a right hemicolectomy (R0-resection), aiming to prevent an ileus. After postoperative recovery a biopsy was taken from a bone lesion of the pelvis, but the histology revealed no malignant tumor cells. Thereafter, another biopsy was done from the mediastinal mass. However, again no malignant involvement could be shown by pathological workup. The findings of the computer tomography and the histology without evidence of adenocarcinoma suggested the existence of a lymphangioma in the mediastinum. As the mediastinal mass resulted in a serious compression of the left lung, we decided to undertake a surgical treatment. The en-bloc surgical resection was successfully and histopathological analysis confirmed the diagnosis of a lymphangiomyoma. In conjunction with the bone lesions the final diagnosis was a generalized lymphangiomatosis and the patient was curatively treated by right hemicolectomy (G2, pT3, pN0 (0/29), pM0). Actually the patient is doing well being in regularly oncological outpatient aftercare. Conclusion: The diagnosis of lymphangiomatosis should be kept in mind even in adult patients, especially in cases with coexistence of cystic mass and osteolytical lesions. Disclosure: No conflict of interest disclosed. P191

Tandem autologous transplantation with melphalane for refractory necrobiotic xanthogranuloma Bittenbring J.1, Pfeifer M.1, Held G.1, Pfreundschuh M.1 UKS Homburg/Saar, Innere Medizin I, Homburg, Germany

1

We report on a 53 year old female treated for necrobiotic xanthogranuloma with tandem autologous stem cell transplantation. Our patient had a IgG-kappa paraprotein, a bone marrow infiltration of 50% plasma cells and anemia of 8,0 g/dl fulfilling the criteria for multiple myeloma alone. On admission she had extensive ulcers around the orbita, on her right shoulder and on the port catheter implantation site which exposed the device on the surface. and on multiple other sites. The ulcers were infected with haemophilus influenzae and pseudomonas aeruginosa. She was diagnosed 8 years ago, treated at several institutions with typical myeloma protocols incorporating mainly dexamethason, bortezomib and lenalidomide. After failure of these treatments she had repeated plasmapheresis for 18 months which stabilized the condition but then the disease progressed again. Background: Necrobiotic xanthogranuloma is a plasma cell dyscrasia and patients develop skin granuloma which progess to necrosis. Treatment is aimed at reducing the paraprotein level. A stem cell transplantation was not yet attempted due to multiple infectious foci and patients wish. However with further progression and unacceptable high disease burden she decided for treatment. The port catheter was removed, a stem cell mobilization with cylcophosphamid was successful with concurrent antibiotic treatment and daily interdisciplinary wound management. High dose melphalan was started immediately thereafter. 14 days later the ulcers began to heal and newer lesions did not appear. At restaging 6 weeks later partial remission was achieved. A second high dose melphalan was performed and the ulcers healed with extensice scarring causing ectropion on both eyes with the need for reconstructive surgery at the ophthalmology departement and LMU Großhadern. Biochemically she is in very good partial remission without evidence for progression of necrobiotic xanthogranuloma 1 1/2 years after autologuous tandem transplantation.

54

Oncol Res Treat 2015;38(suppl 5):1–270

Conclusion: Treatment of necrobiotic xanthogranuloma is aimed at reducing the causative paraprotein and wound healing with combined hematological treatment and surgical & nursing wound management. Disclosure: No conflict of interest disclosed. P192

Pulmonary Marasmius aliaceus infection mimicking aspergillosis Bittenbring J.1, Klotz M.2, Pfeifer M.1, Held G.1, Pfreundschuh M.1 UKS Homburg/Saar, Innere Medizin I, Homburg, Germany, 2UKS Homburg/Saar, Medizinische Mikrobiologie, Homburg/Saar, Germany 1

We report on 32 year old female treated for acute myeloid leukemia (AML with maturation; NPM-1 and FLT3-ITD positive). She received prophylactic posaconazole starting on day 8 after 7+3 induction. In neutropenia on day 18 after induction she had persistent fever > 72 h and a thoracic CT scan showed a round pulmonary nodule in the right upper lobe with halo very suggestive of aspergillosis. Galactomannan screening which was performed twice a week was negative. However treatment for possible aspergillosis was promptly initiated with voriconazole iv. The low-grade fever and infection markers did not resolve the following 10 days. Meanwhile the white blood count resolved and a complete remission of leukemia could be demonstrated. As the aspergillosis did not resolve with normal neutrophil count we switched to liposomal amphothericin B for another 14 days. Radiographic follow up detected cavern formation. Another try with caspofungin resulted in no response for 7 days either. The patients sister was a HLA-match and further delay of consolidative treatment and transplant was not acceptable. An atypical resection of the right upper lobe was performed. As a surprise Marasmius aliaceus (garlic parachute) could be detected by PCR in the surgical specimen. This species was never reported to be a human pathogen. However this could be a reason for the missing response to common antifungals. After quick recovery our patient had a myeloablative conditioning and siblling transplant and is now alive and well on day 250 after transplant. Conclusion: First description of marasmius aliaceus as a human pathogen and a note for the option of surgical treatment of pulmonary fungal infections in selected clinical circumstances. Disclosure: Jörg Bittenbring: Advisory Role: Gilead Michael Pfreundschuh: No conflict of interest disclosed. P193

Adulthood Wiskott-Aldrich-Syndrome Wittke K.1, Hanitsch L.G.1, Grabowski P.1, Volk H.-D.1, Scheibenbogen C.1 Medizinische Immunologie/ Charite Berlin, Immundefektambulanz für Erwachsene, Berlin, Germany 1

Introduction: Wiskott-Aldrich-Syndrome (WAS) is a rare x-linked primary immunodeficiency disease (PID) characterized by the clinical triad thrombocytopenia, ekzema and recurrent infections with an average age at diagnosis younger than two years. Due to advances in the molecular understanding of the disease, improvements in diagnostic tools, supportive care and allogenic hematopoetic cell transplantation (HCT) the morbidity and mortality of WAS could be reduced and more patients reach adulthood. Here we present the case of a 28 year old patient with WAS under supportive therapy. Case: The patient developed petechiae and ekzema as a newborn and had several pulmonary infections during infancy. At the age of 2 years WAS was diagnosed and the patient was taken care of at a specialized pediatric center for PID. Thrombocyte counts fluctuated around 50/nl and he suffered from an Immunoglobuline (Ig)-G- and IgM-deficiency. As the disease-causing genetic alteration a splice site mutation at position +5 of intron 6 of the WAS gene was identified resulting in a truncated WAS protein. In the 1990s the patient’s mother decided against an allogenic HCT. Instead a therapy with IgG was initiated. Under this therapy the patient had minor pulmonary but no life threatening infections. At the age of 24

Abstracts


CONTENTS AUTHOR INDEX

the patient decided to stop IgG therapy and did not see his pediatrician anymore. After two years of increasing rate of infections including at least one pneumonia and worsening ekzema the patient presented to our adult center for PID. After reintroduction of subcutanous IgG therapy, consequent antiinfectious treatment, pneumocystis jiroveci prophylaxis and intensive dermatologic care the patient improved clinically and has been stable with no major infections for almost two years. Discussion: Retrospective studies show a median survival of 14.5 years in patients with classic WAS unless a hematologic and immune reconstitution is reached by allogenic HCT which is today the treatment of choice and is ideally performed under the age of 5 years. The major causes of non HCT associated deaths are infections, bleeding and malignancies. Still there is a growing number of patients reaching adulthood even under supportive therapy only. A smooth transition between pediatric and adult medical care has to be ensured. There are several unanswered questions concerning the optimal medical care for adult WAS patients with virtually no scientific data, e.g. the role of allogenic HCT. Disclosure: No conflict of interest disclosed. P194

Eculizumab therapy for gemcitabine-induced atypical hemolytic uremic syndrome in a patient suffering from adenocarcinoma of the common bile duct: a case report Schneider C.1, Lestin M.1, Diwok C.1, Schneider-Koriath S.1, Steiner R.1, Krammer-Steiner B.1 Klinikum Südstadt Rostock, Rostock, Germany

1

Introduction: Atypical hemolytic syndrome (aHUS) is a rare complication of chemotherapy with gemcitabine presenting with hemolytic anemia, thrombocytopenia, acute kidney failure and arterial hypertension. In most cases aHUS takes a fatal clinical course. We describe the successful treatment of a gemcitabine-induced aHUS with eculizumab, a humanized monoclonal antibody that prevents the production of the terminal complement complex. Methods: A 44 year old male patient suffering from adenocarcinoma of the common bile duct was treated with postsurgical adjuvant chemotherapy with gemcitabine (1000 mg/m² day 1,8,15, new cycle day 29). After the fifth cycle he developed arterial hypertension and a Coombs negative hemolytic anemia. In addition, thrombocytopenia and renal failure with proteinuria occurred. Diagnostic evaluation excluded a gastrointestinal infection and revealed schistocytes in the peripheral blood smear and a normal plasma ADAMTS13 activity. Thus, a gemcitabine-induced aHUS was suggested. Coincidental to the aHUS and complicating the clinical course, the patient developed an adhesive ileus, which had to be surgically revised. Immediately after successful surgery the therapy with corticoids (1g/kg body weight) and plasma exchange was initiated leading to the patient`s clinical stabilisation. Two days later the therapy could be changed to eculizumab (900mg/m² weekly for the first four weeks, thereafter 1200mg/m² every two weeks). Vaccination against Neisseria meningitidis was administered as recommended and accompanying antibiotic prophylaxis was performed for a period of 14 days. Results: Within one week after the first application of eculizumab, the platelet count as the most sensitive parameter of therapy response normalized. Renal function recovered to near normal and proteinuria resolved. Hemodialysis could be prevented. Eventually, under continued eculizumab therapy over the following weeks all blood counts normalized and the patient is now in excellent clinical condition. Arterial hypertension remained and is currently under medical treatment. Despite abandoning the therapy with gemcitabine the patient is still in complete remission. Conclusion: Atypical HUS as a life threatening complication of chemotherapy with gemcitabine was successfully treated with eculizumab. The early adoption of therapy allowed to restore renal function and the prevention of hemodialysis. Disclosure: No conflict of interest disclosed.

Abstracts

P195

Splenomegaly and cytopenia after surgical resection and chemotherapy for colorectal cancer: An interdisciplinary case of hematology and oncology Haas M.1, Rexrodt P.2, Schlitt H.J.3, Vehling-Kaiser U.1 Praxis für Hämatologie, Onkologie und Palliativmedizin, Landshut, Germany, 2Praxis für Radiologie und Strahlentherapie, Landshut, Germany, 3 Universitätsklinikum Regensburg, Klinik und Poliklinik für Chirurgie, Regensburg, Germany 1

Introduction: The current report from an oncologic practice in Germany describes a cross-link between hematology, oncology and surgery Methods: In 2001, a woman then aged 38 years underwent R0 resection for adenocarcinoma of the ascending colon. Afterwards an adjuvant chemotherapy with 5-fluorouracil/folinic acid/oxaliplatin was administered. 4 years later the patient sufferd from relapse with metasis in the right liver lobe and right hemihepatectomy was done. One year later splenomegaly of 18cm x 5cm became evident, but did not cause any symptoms. The blood count then showed a slight tri-cytopenia possibly due to previous chemotherapy. The patient was fully active without any problems due to splenomegaly. In 2013, the patient increasingly suffered from left-sided abdominal pain and fatigue, the spleen then was 20cm in size. Thrombocyte count had dropped below 50 G/L. Bone-marrow puncture did not show signs of dysplastic syndrome or other malignancy. In 2014, the spleen was 23 cm in longest diameter. A CT-scan showed signs of portal hypertension with pronounced collaterals, possibly due to constriction of the inferior vena cava and/or left hepatic vein, possibly due to the liver-resection in 2005. A gastroscopy confirmed oesophageal varices. The patient was transferred to a university hospital for further diagnostics and potential placement of a portosystemic shunt. Left hepatic vein stenosis could not be substantiated by venous catheter pressure measurements. As a definite reason for portal hypertension was not clear with no evidence for liver cirrhosis or fibrosis, the patient was advised for splenectomy for symptom relief. Results: To avoid splenectomy, as preferred by the patient and the treating hemato-oncologist, the case was again discussed with the surgeon. It was concluded to initially perform a side-to-side spleno-renal shunt. After constructing the shunt, a significant reduction of blood-flow through the portal vein directly after surgery was observed. Only three months after the intervention, the spleen had shrunk from 24cm to 16cm accompanied by an increase in thrombocytes above 100 G/L and significant improvement of symptoms. Conclusions: The patient achieved a significant improvement of symptoms after construction of the shunt. Close networking between different disciplines and especially the treating physicians in rural areas and specialists at highly specialized centers were crucial to avoid splenectomy in this case. Disclosure: No conflict of interest disclosed. P196

Chemotherapy and pregnancy: 2 case reports from clinical practice Lestin M.1, Krammer-Steiner B.1 Klinikum Südstadt Rostock, Rostock, Germany

1

Prospectively, the number of pregnant women suffering from cancer will rise due to delayed childbearing and the observed increase in non-Hogdkins lymphoma in the last decades. Oncologists and gynecologists have to face the challenging issue to obtain a good long-term outcome for mother and fetus. Case 1: A 22-year old pregnant woman (25th week of gestation) presented with an inguinal swelling, hepatosplenomegaly, bone lesions, pancytopenia and elevated LDH. Lymph node biopsy revealed diffuse large B-cell lymphoma. We administered 4 courses of standard dose R-CHOP-14. After 3 courses she underwent surgergy because of pathological femur fracture. In 37+2 week of gestation an eutrophic healthy girl was born via

Oncol Res Treat 2015;38(suppl 5):1–270

55


CONTENTS AUTHOR INDEX

normal vaginal delivery. The mother completed therapy with 4 courses R-CHOEP-14 and 4 single doses Rituximab. Case 2: A 33-year old woman presented with breast cancer in 21+0 week of gestation. There was no evidence of metastasis. After breast conservative surgery she received 4 courses adjuvant chemotherapy with epirubicine and cyclophosphamide. In 37+1 week of gestation our patient gave birth to a healthy eutrophic boy via normal vaginal delivery. Her therapy has been completed by 12 courses paclitaxel and radiation. As reported from other authors chemotherapy administered during 2nd and 3rd trimesters of pregnancy seems to be relatively safe. The teratogenicity of chemotherapeutic agents depends on the timing of exposure. Nevertheless chemotherapy during 2nd and 3rd trimesters increases the risk of intrauterine growth restriction. CHOP regimen is considered safe in the 2nd and 3rd trimesters. Rituximab is essential in treatment of aggressive lymphoma. Notably, Rituximab can cross the placenta. As reported in literature neonates exposed to Rituximab presented haematological abnormalities (B-cell depletion, cytopenia) but none had corresponding infections. In pregnant women with breast cancer the decision to recommend mastectomy or breast conservative surgery and also the indication for systemic therapy should follow standard practice. Anthracycline-based regimens remain first choice. Due to risk of fetal complications radiation, trastuzumab and endocrine therapy sholud be initiated after delivery. Disclosure: No conflict of interest disclosed. P197

Simultaneous manifestation of Hodgkin lymphoma and follicular Non-Hodgkin lymphoma Nuber J.1, Fund N.1, Schwella N.1 Klinikum, Ludwigsburg, Germany

1

Introduction: The two main groups of malignant lymphoma are Hodgkin lymphoma (HL) and Non-Hodgkin lymphoma (NHL). In general they are considered as two different entities of malignant lymphoma, but reported cases showed the occurrence of both in the same patient. Most of the studies and case reports describe the development of an NHL after the successful treatment of initially diagnosed HL. This may be related to late treatment-toxicity of chemotherapy and/or radiotherapy. A rare number of cases have shown the possibility of simultaneous manifestation of HL and NHL in the same patient. Especially, the rare occurrence of two histological different lymphomas in the same organ is defined as a composite lymphoma. As the pathogenesis of the occurrence of two histological distinct malignant lymphoma variants in the same patient is not clarified, a possible genetical relation is object of discussion. Case report: A 73-year old male patient was diagnosed with weight loss, night sweat and enlarged axillary and inguinal lymph nodes. The biopsy was performed on one of the two palpable inguinal lymph nodes on the right side. Histological findings showed a classical non-EBV associated HL. Computer tomography showed a splenomegaly without further abdominal and thoracic lymphoma. Furthermore, bone marrow examination showed a 20% infiltration of HL (stage IV B/S). The patient was initially treated with doxorubicin, bleomycin, vinblastine and dacarbazine (ABVD). A follow up computer tomography after 4 courses of ABVD showed in general a partial response. The clinical examination however, surprisingly showed one of the inguinal lymph nodes of constant size, where no biopsy was taken before. Therefore, another biopsy was performed on the non-regressive inguinal lymph node, also located on the right side. The biopsy revealed again Hodgkin and Sternberg-Reed cells, as well as follicular lymphoma cells. After this interesting results we analyzed again the initial biopsy particularly with regard to follicular lymphoma cells, but gained no additional information. This case shows the possible coexistence of a HL and a follicular NHL grade 1/2 already occurring from the beginning on. Conclusion: Although the simultaneous manifestation of HL and NHL in the same patient is rare, this possibility and a second biopsy should

56

Oncol Res Treat 2015;38(suppl 5):1–270

be considered, especially of non- responding areas following treatment against HL. Disclosure: No conflict of interest disclosed. P198

Severe skin complication after implantation of a subcutaneous port system in a patient with leukaemia AML Heißner K.1, Kopp H.-G.1, Ghoreschi K.2, Metzler G.2, Horger M.3, Kanz L.1, Baur R.1, Vogel W.1 Universitätsklinikum Tübingen, Medizinische Klinik 2 Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2Universitätsklinikum Tübingen, Universitäts-Hautklinik, Tübingen, Germany, 3Universitätsklinikum Tübingen, Abteilung für Diagnostische und Interventionelle Radiologie, Tübingen, Germany 1

Case report: A 76-year-old man with acute myeloid leukemia that had evolved from previously diagnosed myelodyspblastic syndrome (MDS) was regularly admitted to our outpatient unit for treatment with 5-Azacytidin s.c. The patient required packed red blood cells on a regular basis and remained chronically neutropenic. Because the patient had lost his right upper arm in early childhood in an accident, repeated left-sided venous access became increasingly difficult. Therefore an implantable venous access device (port) was implanted. The patient was admitted to our emergency department the day after surgery with local erythema, swelling and overheated skin surrounding the scar. The port system was promptly removed and systemic antibiotic treatment was applied. Microbiological smears identified sensitive natural skin flora. Despite excessive use of different antibiotics, reddish-livid skin destruction enlarged with multiple painful ulcerations. Furthermore, distant ulcerations localized around the left elbow emerged. Necrotizing subcutaneous tissue involvement was ruled out by MRI. Deep skin biopsies interestingly showed accumulation of neutrophils despite of persisting peripheral blood °IV neutropenia, consistent with a diagnosis of pyoderma gangraenosum (PG). Treatment with corticosteroids and immune globulins resulted in rapid and ongoing wound healing, confirming the diagnosis. Conclusion: This is a case of PG after implantation of a subcutaneous port devide, resulting in progressive local tissue ulceration not primarily caused by bacterial infection. PG typically occurs in patients with chronic disease, including myelodyplasia. Our case highlights the importance of considering non-infectious causes of severe cutaneous complications after surgical procedures in haematologic patients. PG is a diagnosis of exclusion, skin biopsies are strongly recommended to confirm the diagnosis. Disclosure: No conflict of interest disclosed. P199

Long-lasting remission of metastatic breast cancer with Eribulin Dobbie M.1 Unité d‘Oncologie, Hôpital du Jura, Porrentruy, Switzerland

1

Introduction: Eribulin has been approved to treat patients with metastatic breast cancer who have received at least two prior chemotherapy regimens for late-stage disease, including both anthracycline- and taxane-based chemotherapies. Methods: Case report of a 48 year old patient with metastatic breast cancer treated with Eribulin. Results: This at diagnosis premenopausal patient was diagnosed with ER pos, Her-2 negative, metastatic breast cancer (ulcerated breast lesion with multiple satelite skin metastasis, axillary lymph node metastasis and a malignant pleural effusion) in 2005. Treatment with 6 cycles of palliative chemotherapy with Docetaxel and Epirubicin) from 9/05–1/06 (partial remission). Antihormonal treatment followed with Letrozole from 2/06– 1/07 (stopped because of local progression), Treatment was continued

Abstracts


CONTENTS AUTHOR INDEX

with Tamoxifen with ovarian suppression (recovery of ovarian function) from 1/07–4/12. In 9/2010 pulmonary embolism and heart failure occurred and standard treatment was given with clinical improvement. A right sided tuboovarian abscess (12/10) was treated with antibiotics (patient inoperable) Gynecological follow up showed a growing tumor in the left adnexis and in 2/12 after cardiac recompensation, a staging laparoscopy was performed with left adnexectomy and multiple biopsies of peritoneal implants. The pathological diagnosis was serous adenocarcinoma of the ovary. From 1/12 to 4/12 3 cycles of Carboplatin AUC 5 were given, restaging with CT showed progressive disease. Mechanical ileus developed and right sided adnexectomy with resection of 40 cm of ileum and an ileo-ascendostomy was performed. The second pathological report led to the diagnosis of metastasis of the invasive breast cancer with a typical immunohistochemical profile. After the operation, base line PET/CT showed FDG positive lesions in both breasts, the right axilla, the liver and the right sided peritoneum. Palliative Chemotherapy with Eribulin 1,4 mg/m2 i.v. day 1 and 8, every 3 weeks, was started in 10/12 and the patient is still on treatment after 35 cycles (at time point 5/15). Treatment was well tolerated (no hematological toxicity > grade 1) and the general condition has improved. Follow up radiological examinations show a continous partial remission (in 2/15 no signs of metastasis in lung or liver). Conclusions: The patient tolerated Eribulin very well despite the important comorbidities with an excellent effect on progression free survival.

stage, tumor grade, tumor location, nodal status and FOXP3+ Treg content only pT (p < 0.0001) and pN (p < 0.0001) had significance, but not tumor grade (p = 0.42), tumor localization (p = 0.1) or FOXP3+ Treg infiltration (p = 0.07). Conclusion: FOXP3+ Treg cell density in CRC may help as a predictive biomarker for the risk assessment and therapy management of early and middle stage colorectal cancer.

Disclosure: Michael Dobbie: Employment or Leadership Position: Leitender Arzt Onkologie, Hôpital du Jura; Financing of Scientific Research: Eisei.

Introduction: In the CORRECT Ph3 trial (NCT01103323), regorafenib improved overall survival (OS) and progression-free survival (PFS) vs placebo in patients with mCRC who progressed on standard therapies (Grothey 2013). Initial biomarker analyses suggested that regorafenib provides a clinical benefit in various mutational subgroups (Jeffers 2013). Here we present additional exploratory analyses to evaluate clinical benefit in CRC subgroups defined by gene expression. Methods: Gene expression analysis (Affymetrix ST1.0 array) was conducted on archival tumor tissue from 281 of the 760 patients (37%) enrolled in CORRECT. Next-generation sequencing (NGS) was done on 239 specimens (FoundationONE). Gene expression data were subjected to hierarchical molecular tumor classification (Marisa 2013). Cox proportional hazards models were used to identify potential prognostic or predictive biomarkers. Results: The distribution of the 6 different CRC subtypes characterized by gene expression clusters originally defined by Marisa and the classification of the 281 patients from CORRECT are shown in the table.

Posterdiskussion

Kolon-/Rektumkarzinom P200

High intratumoral FOXP3+ T regulatory cells (Tregs) density in colorectal cancer is associated with improved survival but unrelated to tumor localization Marx A.H.1, Sauter G.1, Simon R.1, Bokemeyer C.2, Terracciano L.3, Izbicki J.R.4, Melling N.4 University Medical Center Hamburg-Eppendorf, Hamburg, Pathology, Hamburg, Germany, 2Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany, 3University Hospital Basel, Pathology, Basel, Switzerland, 4University Medical Center Hamburg-Eppendorf, Hamburg, Surgery, Hamburg, Germany 1

Introduction: To determine the density and prognostic significance of FOXP3+ T regulatory cells (Tregs) in colorectal cancer of different localizations (right versus left colon) compared with conventional histopathologic features and with CD3+ and CD8+ lymphocyte densities. Methods: Tissue microarrays (TMA) and immunohistochemistry (IHC) were used to examine the densities of CD3+, CD8+ and FOXP3+ lymphocytes in normal colonic mucosa and tumor tissue from 1800 colorectal cancers. These densities were evaluated for associations with histopathologic features and patient survival. Results: FOXP3+ Tregs density was low in 547 (50.5%), high in 120 cases (11.1%). FOXP3+ Tregs density was associated with a high CD8+ (p = 0.018) and CD3+ density (p < 0.0001) in tumor tissue. FOXP3+ Tregs density was higher in tumor tissue compared to normal tissue (p < 0.0001). Low FOXP3+ Tregs density was significantly associated with high tumor grade (p < 0.0001), high tumor stage (p < 0.0001) and nodal status (p = 0.023), but there was no correlation to histological subtypes (mucinous, signet cell; p = 0.42) or tumor localization (right versus left; p = 0.17). High density of FOXP3+ Tregs, CD3+ and CD8+ cells in tumor tissue was related to a better patient survival (p < 0.0001, p = 0.02 and p = 0.01 respectively; Figure 3). In a multivariate analysis including tumor

Abstracts

Disclosure: No conflict of interest disclosed. P201

Molecular subtypes and outcomes in regorafenib-treated patients with metastatic colorectal cancer (mCRC) enrolled in the CORRECT trial Karthaus M.1, Schwenke S.2, Seidel H.3, Beckmann G.2, Reischl J.2, Vonk R.2, Lenz H.-J.4, Tebernero J.5, Siena S.6, Grothey A.7, van Cutsem E.8, Jeffers M.9, Wagner A.2, Laurent D.2, Kobina S.9, Rutstein M.9, Guinney J.10, Tejpar S.11 Klinikum Harlaching, Klinik für Hämatologie, Onkologie und Palliativmedizin, München, Germany, 2Bayer Pharma AG, Berlin, Germany, 3Bayer HealthCare Pharmaceuticals, Berlin, Germany, 4University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, United States, 5Vall d’Hebron University Hospital, Barcelona, Spain, 6Ospedale Niguarda Ca’ Granda, Milano, Italy, 7Mayo Clinic, Rochester, United States, 8University Hospitals Gasthuisberg, Leuven, Belgium, 9Bayer HealthCare Pharmaceuticals, Whippany, United States, 10 Sage Bionetworks, Seattle, United States, 11Molecular Digestive Oncology, Leuven, Belgium 1

Tab. 1. Distribution of molecular subtypes

Subtype

CINImmuneDown (C1)

dMMR (C2)

KRASmutant (C3)

CSC (C4)

CINWntUp (C5)

CINnormL (C6)

Marisa (2013)

21%

19%

13%

10%

27%

10%

CORRECT (n)

36% (100)

12% (33)

11% (32)

7% (19)

32% (90)

2% (7)

The six Marisa subtypes derived a similar OS benefit from regorafenib. Although the numbers of patients in the subgroups are small, a greater PFS benefit for regorafenib was seen in patients in the ‘high-risk’ subgroup (C4 + C6, n = 26; HR = 0.10; 95% CI 0.02–0.35; p = 0.0009) than the ‘lowrisk’ subgroup (C1+C2+C3+C5, n = 255; HR = 0.58; 95%CI 0.44–0.77; p = 0.002). NGS analyses suggested that the chromosomal instability group (CIN) was the predominant subgroup in CORRECT. Conclusion: Molecular characterization by gene expression analysis may allow the identification of CRC subgroups that correlate with regorafenib clinical benefit. Data suggest this does not depend on the presence of mutations. Additional exploratory analyses, including the use of the recently

Oncol Res Treat 2015;38(suppl 5):1–270

57


CONTENTS AUTHOR INDEX

defined Consensus molecular subtypes (Dienstmann ASCO 2014), to validate these results are ongoing. Disclosure: Meinolf Karthaus: Expert Testimony: Bayer Sabine Tejpar: No conflict of interest disclosed.

Gebauer L.1, Stabla K.1, Nist A.2, Mernberger M.2, Wischmann V.3, Stiewe T.2, Barckhausen C.1, Moll R.3, Brendel C.1, Neubauer A.1 Philipps University of Marburg and University Hospital of Giessen and Marburg, Department of Hematology, Oncology and Immunology, Marburg, Germany, 2 Philipps University of Marburg, Molecular Oncology, Marburg, Germany, 3 Philipps University of Marburg, Institute of Pathology, Germany 1

P202

Molecular targeting of EGFR-downstream effectors in colorectal cancer cells by antibody-mediated siRNA treatment Appel N.1, Bäumer N.1, Wardelmann E.2, Buchholz F.3, Müller-Tidow C.4, Berdel W.E.1, Bäumer S.1 Universitätsklinikum Münster, Medizinische Klinik A, Labor für Molekulare Hämatologie und Onkologie, Münster, Germany, 2Universitätsklinikum Münster, Gerhard-Domagk Institut für Pathologie, Münster, Germany, 3TU Dresden, UCC, Medical Systems Biology, Medizinische Fakultät, Dresden, Germany, 4 Universitätsklinikum Halle, Innere Medizin IV, Hämatologie und Onkologie, Halle, Germany 1

Introduction: RAS-mutated carcinomas are resistant against EGFR-antibody therapies such as Cetuximab. We overcame this therapy resistance in a proof-of-principle study ADDIN RW.CITE{{168 Bäumer,S. 2015}}[1]. In this study, we coupled the anti-EGFR-antibody Cetuximab chemically to protamine via the linker sulfo-SMCC. This complex carries siRNA into EGFR-positive cells such as the colorectal cancer cells lines HCT116, LoVo, DLD-1, SW480 and HT29. The siRNA-mediated downregulation of KRAS leads to silencing of MAPK pathway signal transduction, decreased proliferation and enhanced apopotosis. Since tumor growth in vivo was inhibited but still detectable, we combined KRAS downregulation with siRNA treatments against additional EGFR-downstream factors that might be operative in MAPK independent tumor cell growth. Methods: We used the Cetuximab-sulfo-SMCC-protamine complex to bind siRNAs e.g. against PI3CA, which is mutated or overexpressed in many tumors. We treated colorectal cancer cell lines with KRAS-siRNA-, PI3CA-siRNA- or combined Cetuximab-complexes. Cells were then subjected to colony formation assays in soft agar, MTS assays to determine vitality and proliferation. Also, we injected cells subcutaneously into the flank of CD1-nude mice and treated these mice with the Cetuximab-siRNA complexes. Results: Downregulation of PI3CA in colorectal cancer cells leads to significantly decreased colony formation. Moreover, combination of siRNA against KRAS and PI3CA coupled to Cetuximab-complexes dramatically slowed down tumor growth in vivo. Conclusions: Antibody-mediated knockdown of KRAS in KRAS-mutated tumors restores EGFR-sensitivity. Downregulation of additional signaling pathways leads to a cooperative treatment success. This indicates that complete molecular characterization of multiple signaling pathways driving malignant behaviour of the respective cell type enables us to tailor the siRNA delivery system to the specifically expressed oncogenes and consequently in a given cell line or tumor yields optimal therapeutic activity. Reference: 1 Bäumer S, Bäumer N, Appel N, et al.: Antibody-mediated delivery of anti-KRAS-siRNA in vivo overcomes therapy resistance in colon cancer. Clin Cancer Res 2015;21:1383–1389. Disclosure: No conflict of interest disclosed.

58

P203

Colorectal carcinoma patients with wild-type PIK3CA tumors show improved overall survival

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: Specific gene mutations can influence the response of tumors to anticancer drugs. Thus, molecular methods gain in importance for clinical diagnostics to create a decisional base for the administration of a given drug. In the past, there have been conflicting results with regard to aspirin use and survival in correlation with PIK3CA mutations in colorectal cancer patients. This retrospective study was done to understand a potential relation between PIK3CA mutation status and aspirin use in colorectal cancer. Patients and methods: This was a retrospective analysis of paraffin-embedded tumor tissue of 153 patients suffering from colorectal carcinoma stage I-III (UICC classification) or IV with only a few resectable liver metastases. Genomic DNA was subjected to analysis of PIK3CA and KRAS mutation status by pyrosequencing or semiconductor sequencing, respectively. Mutation status and regular aspirin use were correlated to 5-year and 10-year overall survival (OS). Results: With regard to the entire patient cohort, aspirin use per se did not affect 10-year-OS significantly. However, wild-type PIK3CA status was associated with a significantly higher rate of 10-year OS (48% vs. 21%, p = 0.01, Fisher`s exact test). Detailed subgroup analyses revealed that especially aspirin users with wild-type PIK3CA tumors show significantly improved 5-year OS (75% vs. 25%, p = 0.01, Fisher’s exact test) and 10year OS (57% vs. 13%, p = 0.05, Fisher’s exact test). KRAS mutation did not impact OS in our CRC patient cohort. Conclusions: Our data indicate that post-diagnosis aspirin intake could be beneficial especially for aspirin users with PIK3CA mutation while KRAS status is irrelevant for clinical diagnostics with regard to a decision for or against aspirin prescription. Disclosure: No conflict of interest disclosed. P204

Comparison of therapeutic efficacy of single VEGF- versus combined VEGF/Angiopoietin-2 inhibition in combination with chemotherapy in a colorectal carcinoma xenograft model Freystein J.1, Reipsch F.1, Caysa H.1, Schmoll H.-J.2, Müller-Tidow C.1, Müller T.1 Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik für Innere Medizin IV, Hämatologie/Onkologie, Halle (Saale), Germany, 2Martin-LutherUniversität Halle-Wittenberg, AG Klinische Studien in der Onkologie, Halle (Saale), Germany 1

VEGF inhibition by addition of Bevacizumab to chemotherapy regimen of metastatic colorectal cancer leads to improved outcome. However, anti-angiogenic tumor therapy targeting a single factor may be limited by complementary mechanisms. It was shown that high Angiopoietin-2 (Ang-2) serum levels are associated with poor clinical outcome of colorectal cancer patients treated with Bevacizumab-containing therapy. The aim of the present study was to compare therapeutic efficacy of single VEGFvs. combined VEGF/Ang-2 inhibition using a novel bi-specific antibody (CrossMab) in combination with chemotherapy in a colorectal cancer xenograft model. Subcutaneous xenograft tumors were generated in athymic nude mice using the human colorectal cancer cell line DLD-1. Monitoring of tumor growth was performed by caliper measurement. Mean tumor volume per group (n = 10) at start of treatment was around 130 mm3. Chemotherapy consisted of a combination of 5-fluorouracil and irinotecan. Following antibodies provided by Roche were used: anti-VEGF (B20.4.1), CrossMab

Abstracts


CONTENTS AUTHOR INDEX

(B20.4.1/LC06). Single and combination treatments were performed by i.p. injections once weekly. Mean tumor volume of control group reached 1400 mm3 on d18 after start of treatment with 4 of 10 tumors exceeded 1500 mm3 and treatment was stopped. Chemotherapy and anti-VEGF treatment inhibited tumor growth to some extent and had to be completed on d27 when 4 tumors within each group exceeded 1500 mm3. An efficient tumor growth inhibition could be achieved by single treatment with the CrossMab and both combination regimen. The study was completed on d49 when 2 tumors within the CrossMab- and anti-VEGF/chemotherapy groups had exceeded 1500 mm3. Mean tumor volumes at this time point were 1018 mm3, 829 mm3 and 296 mm3 for anti-VEGF/chemotherapy, CrossMab and CrossMab/chemotherapy, respectively. Analysis of single tumors revealed that 3 of 10 tumors in the anti-VEGF/chemotherapy group, 5 of 10 tumors in the CrossMab group and 10 of 10 tumors in the CrossMab/chemotherapy group had shown complete growth retardations. Statistical analysis on d18 showed that CrossMab alone and both combination treatments led to a significant inhibition of tumor growth compared to PBS control. The difference between CrossMab/chemotherapy and anti-VEGF/chemotherapy on d49 was highly significant. In conclusion, the therapy regimen comprising combined VEGF/Ang-2 inhibition was clearly superior to combination therapy with single VEGF inhibition. Disclosure: Juana Freystein: Expert Testimony: Die Studie wurde durch Drittmittel der Firma Roche unterstützt. Thomas Müller: Expert Testimony: Die Studie wurde durch Drittmittel der Firma Roche unterstützt. P205

Heterogeneity of colorectal cancer liver metastasis – analysis of WNT pathway patterns via High-throughput and bioinformatics profiling Bleckmann A.1,2, Wachter A.2, Conradi L.-C.3, Wolff A.2, Hoppenau C.4, Korf U.5, Schildhaus H.-U.4, Homayounfar K.3, Pukrop T.1, Beissbarth T.2 Dept. of Hematology and Medical Oncology, Göttingen, Germany, 2Dept. of Medical Statistics, Göttingen, Germany, 3Dept. of General, Visceral and Pediatric Surgery, Göttingen, Germany, 4Dept. of Pathology, Göttingen, Germany, 5 German Cancer Research Center (DKFZ), Heidelberg, Germany 1

Introduction: In cancer progression the development of distant metastases is the crucial adverse event. Focusing on colorectal cancer, the majority (70%) of patients develops metastasis in the liver. Within the BMBF-founded consortium MetastaSys the role of the mutational activation in the WNT pathway are investigated in these metastases. In order to address the phenomenon of intermetastatic heterogeneity the established metastasis profiling was applied to several metastases of the same patient. Methods: Two pilot cases including four and two liver metastases, respectively as well as corresponding normal liver tissue were comprehensively characterized in terms of clinical annotations as well as standardized histopathological and molecular subtyping. All tumor samples were subtyped according to the specific mutational status of KRAS, NRAS, BRAF and PIK3CA as well as alterations in DNA mismatch repair enzymes and microsatellite instability. Fresh frozen samples for transcriptome analyses were characterized by an experienced pathologist (tumor cell content, amount of stroma, inflammatory infiltrate and necrosis) to assure high quality of tissue and to obtain valid data. After characterization, samples were profiled via subsequent transcriptome sequencing and proteomics profiling via Reverse-Phase-Protein-Arrays (RPPA). Results: To access the clinical and genetical characterization of the samples a standardized data analysis pipeline for processing the RNA-Seq data was established. The principle-component-analysis revealed a good separation between normal tissue and tumor samples. Interestingly, the correlation between healthy samples among different patients was higher than between the metastatic samples of the same patient. Furthermore, relevant WNT markers such as AXIN2, Lef1 and CDX2 were within the top 25 upregulated genes discriminating between normal and metastatic samples.

Abstracts

Conclusion: RNA-Seq analysis allows a clear discrimination of normal and metastatic liver tissue. In different metastases of the same patient heterogeneous expression patterns in the WNT pathway are identified with relevant implication for clinical treatment approaches. Based on these findings and the positive validation of the established methodology including innovative High-throughput profiling and comprehensive bioinformatics approaches we started analyzing a large number of metastatic patient samples within the consortium. Disclosure: No conflict of interest disclosed. P206

Loss of H2Bub1 expression is linked to poor prognosis in nodal negative colorectal cancers Marx A.H.1, Simon R.1, Bokemeyer C.2, Sauter G.1, Terracciano L.3, Izbicki J.R.4, Melling N.4 University Medical Center Hamburg-Eppendorf, Pathology, Hamburg, Germany, 2Hubertus Wald Cancer Center, University Medical Center HamburgEppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany, 3University Hospital Basel, Pathology, Basel, Switzerland, 4University Medical Center Hamburg-Eppendorf, Surgery, Hamburg, Germany 1

Introduction: To correlate H2Bub1 expression with outcome in colorectal cancer. Methods: H2Bub1 expression was analyzed by immunohistochemistry on a tissue microarray containing 1800 colorectal cancers. Results were compared to clinicopathological parameters. Results: H2Bub1 IHC was seen in 1256 (79.3%) of 1584 interpretable CRC and was considered weak in 26.2% and strong in 53.1% of cancers. H2Bub1 expression was completely lost in 20.7% of the cases. Loss of H2Bub1 expression was associated with high tumor grade (p = 0.0211), high tumor stage (p = 0.0003), positive nodal status (p = 0.0139) and histological tumor type (p = 0.0202). No link was found between H2Bub1 expression and tumor localization (p = 0.1262), peritumoral lymphocytic infiltration (p = 0.2523) or vascular invasion (p = 0.5970). Loss of H2Bub1 expression in CRC was strongly associated with poor patient survival (p = 0.0006). This observation held true also in a subset survival analysis of nodal negative (N0) and nodal positive (N1) cancers (p = 0.0296 and p = 0.0197, respectively). In the subgroup of p53 negative cancers no prognostic impact of H2Bub1 staining was seen (p = 0.1924), whereas in p53 positive CRC H2Bub1 expression loss was associated with poor prognosis (p = 0.0031). Strikingly worsened outcome was found for nodal negative cancers presenting with accumulation of p53 when H2Bub1 expression was lost (p = 0.0006). Conclusion: Our data demonstrate that a reduced H2Bub1 expression is a strong prognostic biomarker both in nodal negative and nodal positive CRC. H2Bub1 expression measurement might help to select nodal negative CRC patients that may benefit from adjuvant therapy. Disclosure: No conflict of interest disclosed. P207

High Ki67 expression in colorectal cancer is an independent prognostic marker associated with good prognosis and leftsided tumor localization Marx A.H.1, Simon R.1, Bokemeyer C.2, Terracciano L.3, Simon G.1, Izbicki J.R.4, Melling N.4 University Medical Center Hamburg-Eppendorf, Pathology, Hamburg, Germany, 2Hubertus Wald Cancer Center, University Medical Center HamburgEppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany, 3University Hospital Basel, Pathology, Basel, Switzerland, 4University Medical Center Hamburg-Eppendorf, Surgery, Hamburg, Germany 1

Introduction: To correlate Ki67 expression with outcome and other molecular parameter in colorectal cancer.

Oncol Res Treat 2015;38(suppl 5):1–270

59


CONTENTS AUTHOR INDEX

Methods: Ki67 labeling index (Ki67LI) was analyzed by immunohistochemistry on a tissue microarray containing 1800 colorectal cancers. Results were compared to clinicopathological and molecular parameters. Results: Ki67LI was considered low in 26.3%, moderate in 56.7% and high in 17.0% of 1653 interpretable CRCs. High Ki67 expression was associated with low tumor stage (p < 0.0001) and left-sided tumor localization (p = 0.0280), but not with tumor grade (p = 0.8948), nodal status (p = 0.0702) or histological tumor type (p = 0.7274) and was an independent prognosticator of favorable survival (p = 0.0004). High Ki67 expression was also significantly linked to high-level nuclear β-catenin and p53 expression (p < 0.0001 and p = 0.0288, respectively). Conclusion: Our data show that high Ki67 expression in colorectal cancers is associated with good clinical outcome and has a preference for left-sided tumors. Ki67, p53 and β-catenin overexpression seem to be linked in CRC and indicate a cellular state of high proliferative activity. Finally, our findings strongly argue for a clinical utility of Ki67 immunostaining as an independent prognostic biomarker in colorectal cancer enabling to select patients for adjuvant treatment and contributing to the prognostic evaluation in these patients. Disclosure: No conflict of interest disclosed. P208

Expression of phospho-mTOR kinase is abundant in colorectal cancer and associated with left-sided tumor localization – a potential therapy target? Marx A.H.1, Simon R.1, Izbicki J.R.2, Bokemeyer C.3, Terracciano L.4, Sauter G.1, Melling N.2 University Medical Center Hamburg-Eppendorf, Pathology, Hamburg, Germany, 2University Medical Center Hamburg-Eppendorf, Surgery, Hamburg, Germany, 3Hubertus Wald Cancer Center, University Medical Center HamburgEppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany, 4University Hospital Basel, Pathology, Basel, Switzerland

P209

RBM3 expression loss is associated with right-sided localization and poor prognosis in colorectal cancer Marx A.H.1, Simon R.1, Izbicki J.R.2, Terracciano L.3, Bokemeyer C.4, Sauter G.1, Melling N.2 University Medical Center Hamburg-Eppendorf, Pathology, Hamburg, Germany, 2University Medical Center Hamburg-Eppendorf, Surgery, Hamburg, Germany, 3University Hospital Basel, Pathology, Basel, Switzerland, 4Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany 1

Introduction: The RNA-binding motif protein 3 (RBM3) has recently been suggested as a prognostic biomarker in an array of human cancers. Methods: RBM3 expression was analyzed by immunohistochemistry on a tissue microarray containing 1800 colorectal cancers (CRC). Results: Nuclear RBM3 immunohistochemical staining was found in 95.9% of all interpretable colorectal cancers. Loss of RBM3 expression was linked to advanced tumor stage (p < 0.0001), right-sided tumor localization (p < 0.0001) and poor prognosis (p = 0.0003). In a multivariate analysis, including RBM3 staining, tumor grade, tumor stage and nodal status, only tumor stage and nodal status proved to be independent prognostic markers (p < 0.0001 each), whereas the prognostic impact of RBM3 staining was not significant (p = 0.2655). Conclusion: Our observations indicate that loss of RBM3 expression is an unfavorable prognostic marker in CRC and linked to right-sided tumor localization. Disclosure: No conflict of interest disclosed.

1

P210

βIII-tubulin overexpression is linked to left-sided tumor localization and nuclear β-catenin expression in colorectal cancer

Introduction: The mammalian target of rapamycin (mTOR) has been suggested as a prognostic biomarker and therapeutic target in an array of human cancers. Methods: phospho-mTOR (p-mTOR) expression was analyzed by immunohistochemistry (IHC) on a tissue microarray containing 1800 colorectal cancers (CRC). Clinical follow-up data were available from all cancer patients. Results: Positive p-mTOR immunostaining was seen in 83.5% of 1640 interpretable CRC and was considered weak in 862 (52.5%) and strong in 508 cases (31.0%). Matching clinico-pathological parameters were available in 1580 cases. p-mTOR staining was more frequent in tubular adenocarcinomas than in the less common histological subtypes (mucinous, medullary, signet cell; p = 0.0163) and significantly linked to carcinomas of the left-sided colon and rectum as compared to right-sided CRC (p = 0.0066). There was no significant association between p-mTOR expression and patients’ gender, tumor stage, tumor grade or nodal status. In a survival analysis, p-mTOR IHC status of all CRC was unrelated to patient survival (p = 0.702). In a multivariate analysis including pT, pN, tumor grade, tumor localization and p-mTOR expression, only pT, pN (both p < 0.0001) and grade (p = 0.0001) showed prognostic impact, but not tumor localization (p = 0.9472) or p-mTOR expression (p = 0.8879). Conclusion: Our observations indicate that p-mTOR overexpression is abundant in CRC, and linked to left-sided tumor localization. The high frequency and overexpression of p-mTOR is providing further rationale for targeting this pathway therapeutically in CRC patients. However, a prognostic role of p-mTOR overexpression in CRC could not be confirmed.

Marx A.H.1, Simon R.1, Bokemeyer C.2, Terracciano L.3, Sauter G.1, Izbicki J.R.4, Melling N.4

Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

60

Oncol Res Treat 2015;38(suppl 5):1–270

University Medical Center Hamburg-Eppendorf, Pathology, Hamburg, Germany, 2Hubertus Wald Cancer Center, University Medical Center HamburgEppendorf, Department of Oncology, Hematology, BMT with section Pneumology, Hamburg, Germany, 3University Hospital Basel, Pathology, Basel, Switzerland, 4University Medical Center Hamburg-Eppendorf, Surgery, Hamburg, Germany 1

Introduction: βIII-tubulin expression correlates with poor outcome in various malignancies. Materials and methods: βIII-tubulin expression was analyzed by immunohistochemistry on a tissue microarray containing 1800 colorectal cancers. Results were compared to clinic-pathological and molecular parameters. Results: βIII-tubulin expression was detectable in 79.2% of 1619 interpretable colorectal cancers. High βIII-tubulin expression was associated with left-sided tumor localization (p = 0.0303) and nuclear β-catenin expression (p = 0.003). High βIII-tubulin expression was not linked to the gender of the patient (p = 0.5842). When all tumors were analyzed the prognostic role of βIII-tubulin expression was not independent of pT stage, pN stage, tumor grade or tumor localization (p = 0.0517). Conclusion: βIII-tubulin expression is not an independent prognostic parameter in colorectal cancer. The significant association with left-sided tumor localization and a key genomic alteration of colorectal cancer such as β-catenin suggest interaction with important pathways involved in colorectal cancer.

Abstracts


CONTENTS AUTHOR INDEX

P211

P212

Subgroup analysis of patients with metastatic colorectal cancer (mCRC) treated with regorafenib (REG) in the CORRECT trial who had progressionfree survival (PFS) longer than 4 months

Tp53-dependent response to chemo- and radiotherapy in colorectal cancer (CRC) cell lines

Stein A.1, Grothey A.2, Sobrero A.3, Siena S.4, Falcone A.5, Ychou M.6, Humblet Y.7, Bouché O.8, Mineur L.9, Barone C.10, Adenis A.11, Tabernero J.12, Yoshino T.13, Lenz H.-J.14, Goldberg R.M.15, Xu L.16, Wagner A.17, van Cutsem E.18 Universitätsklinikum Hamburg-Eppendorf, II. Medizinische Klinik und Poliklinik, Hamburg, Germany, 2Mayo Clinic, Rochester, United States, 3San Martino Hospital, Genova, Italy, 4Ospedale Niguarda Ca’ Granda, Milan, Italy, 5 Ospedaliero-Universitaria Pisana, Pisa, Italy, 6ICM Val d’Aurelle, Montpellier, France, 7Cliniques Universitaires Saint-Luc, Brussels, Belgium, 8Centre Hospitalier Universitaire Robert Debré, Reims, France, 9Institut Sainte-Catherine, Avignon, France, 10Catholic University of Sacred Heart, Roma, Italy, 11Centre Oscar Lambret, Lille, France, 12Vall d’Hebron University Hospital, Barcelona, Spain, 13 National Cancer Center Hospital East, Kashiwa, Japan, 14University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, United States, 15 Ohio State University Comprehensive Cancer Center, Columbus, United States, 16 Bayer HealthCare Pharmaceuticals, Whippany, United States, 17Bayer Pharma AG, Berlin, Germany, 18University Hospital Gasthuisberg, Leuven, Belgium 1

Introduction: In the CORRECT phase III trial (NCT01103323), the multikinase inhibitor REG significantly improved overall survival (OS) and PFS vs placebo in patients with mCRC who had disease progression after other standard therapies (HR for OS: 0.77; 1-sided p = 0.0052; Grothey, 2013). A post hoc exploratory subgroup analysis was conducted on patients in the REG treatment group who had a PFS longer than 4 months (long PFS). Methods: PFS was defined as the time from randomization to disease progression (radiological or clinical) or death from any cause (whichever occurs earlier). Of the 505 patients randomized to REG in CORRECT, 98 (19%) were classified as having long PFS benefit, defined as a PFS event >4 months after randomization. Baseline patient characteristics, safety, and dosing parameters were analyzed. Results: The long PFS subpopulation was representative of the overall REG-treated population (table). Long PFS patients received a median of 6 cycles of REG (range 1–12); 92% received ≥5 cycles, and 20% had >8 cycles. Adverse events (AEs) of any grade were experienced by all long PFS patients, and the most common grade ≥3 AEs were hand-foot skin reaction (20%), hypertension (17%), diarrhea (17%), and fatigue (16%). Conclusion: A subset of 98 (19%) patients treated with REG in the CORRECT study had a PFS >4 months, confirming the clinical benefit and tolerability of REG as a treatment option for patients with mCRC. Patients with long PFS generally had better ECOG PS and fewer tumor sites than the overall population. Prospective validation of these findings in conjunction with biomarker analysis from real-life clinical experience is needed. Tab. 1. Baseline characteristics

All patients (n = 505)

Long-PFS (n = 98)

Median age, yrs (range)

61 (22–82)

61 (34–82)

ECOG PS,%

0/1

52/48

63/37

Primary tumor,%

Colon/ Rectum

64/30

52/37

Tumor sites,%

1/2/3

19/36/27

30/38/16

KRAS status,%

Mutant/ Wild-type

54/41

47/44

Disclosure: Alexander Stein: Financing of Scientific Research: Bayer, Roche, ­Sanofi, Merck Serono, Amgen; Expert Testimony: Roche, Sanofi. Eric van Cutsem: Expert Testimony: Bayer.

Abstracts

Milazzo A.1, Kanz L.1, Schittenhelm M.M.1, Kampa-Schittenhelm K.1 Universitätsklinikum, Tübingen, Germany

1

Background: Colorectal cancer is one of the most common malignancies with high prevalence and low 5-year survival in the advanced stages. Inactivating mutations of the tp53 gene are associated with resistance to therapy in several tumor models, conferring poor prognosis. In CRC, where tp53 mutations occur in nearly half of the cases, the role of tp53 remains unclear. The objective of our study was to evaluate the role of tp53 signaling for response to standard chemotherapy. In addition, we tested whether combination approaches with radiotherapy might overcome therapy resistance in mutant-tp53 CRC – thus potentially providing a novel molecular-defined therapeutic rationale in locally advanced disease. Methods: Three independent isogenic CRC cell line models (HCT116, RKO, DLD-1), harboring a genetically modified tp53-null variant or tp53 wild type (wt) were generously provided by Dr. Vogelstein (JHU, Baltimore, MD). Cells were treated with 5-Fluoruracil (5-Fu), Oxaliplatin (Ox) or Irinotecan (Ir) with escalating doses and/or gamma irradiation (5Gy) and effects on cellular proliferation, viability, induction of apoptosis and cell cycle were assessed. Results: We show, that proapoptotic effects mediated via 5-Fu, IR and Ox depend on tp53: significantly higher rates of proapoptotic cells were observed in a dose-dependent manner for the tp53 wt cell strains compared to the corresponding tp53 null cells (with the exception of Ir in DLD-1 cells). In contrast, exposure of tp53 wt vs. tp53 null cell strains tp gamma irradiation at 5 Gy did reveal equipotent p53-independent induction of apoptosis – providing the rational to combine radiation therapy with chemotherapy in mutant-tp53 CRC. Indeed and in particular the tp53 null strains benefited when radiotherapy was combined with either of the chemotherapeutics. Importantly, in the tp53 wt cell strains, we observed adverse effects: Consistent for all wt cell lines, addition of radiotherapy to either agent decreased the proapoptotic effect seen for chemotherapy alone – partly due to unfavorable cell cycle arrest phenomena. Conclusion: Our findings demonstrate a role of intact p53 signaling for the efficacy of Ox, IR and 5Fu in CRC models. Radiochemotherapy is only of benefit in mutant-tp53 cancer, which provides a rationale for tp53 mutation screening as a helpful tool for assessment of therapeutic options. Disclosure: No conflict of interest disclosed. P213

Analysis of the quality of life in patients treated with Aflibercept and FOLFIRI for metastatic colorectal cancer (mCRC) – Interim results from the non-interventional QoLiTrap study Derigs H.-G.1, Scholten F.1, Losem C.2, Kröning H.3, Windemuth-Kieselbach C.4, Hofheinz R.-D.5 Klinikum Frankfurt Höchst, Klinik für Innere Medizin 3, Frankfurt a. M., Germany, Johanna-Etienne Krankenhaus, Praxis für Hämatologie und Onkologie, Neuss, Germany, 3Schwerpunktpraxis für Hämatologie und Onkologie, Magdeburg, Germany, 4Alcedis GmbH, Gießen, Germany, 5Universitätsmedizin Mannheim, Interdisziplinäres Tumorzentrum, Mannheim, Germany 1 2

Introduction: Aflibercept is an anti-angiogenic fusion protein targeting VEGF-A, VEGF-B and PlGF. It is approved in combination with FOLFIRI for patients with mCRC, who have already received an Oxaliplatin containing therapy. Methods: Patients with mCRC receiving Aflibercept according to label are included in QoLiTrap, an ongoing non-interventional study to evaluate quality-of-life in patients (AIO-LQ-0113). The study aims to enroll 1500 patients in Germany, Austria and Switzerland. The primary aim is to assess health related quality of life in clinical practice. EORTC-QLQ C30 questionnaires are filled in by the patients at baseline and in every other cycle.

Oncol Res Treat 2015;38(suppl 5):1–270

61


CONTENTS AUTHOR INDEX

Results: This is the interim analysis of the first 348 included patients who have received at least one cycle of Aflibercept. 329 patients have completed EORTC-QLQ C30 at baseline and 218 patients completed at least baseline and 2 additional questionnaires. These 218 patients received Aflibercept therapy in 1th (10%), 2nd (48%), 3rd (19%) and later lines (22%) with a median of 6 (and up to 35) cycles. Median age was 66 years with good PS (ECOG 0: 38%; ECOG 1: 45%). Complete or partial remission was observed in 12% of all evaluable patients over all lines, SD in 27% and PD in 14% (response not yet documented for 47%). At baseline, patients presented a median EORTC global health score of 58.3 (n = 213). Differences between baseline and mean values after 12 weeks were observed in global health status as well as in physical-, emotional-, cognitive-, role- and social functioning. Changes in physical functionality were mainly driven by an increase in fatigue, whereas in other symptom scales such as constipation and financial impact the value declined. This first analysis from the QoLiTrap study has identified no new safety signals. Conclusion: QoLiTrap is a large observational study conducted to analyze the quality of life in patients with mCRC treated with FOLFIRI & Aflibercept. This interim analysis shows preliminary, yet encouraging results with PR or SD adding to a tumor control rate of 74% within the cohort of patients with response documentation. The decline in global health status was moderate. Updated results will be presented at the meeting. This study is supported by Sanofi. Disclosure: Hans-Günter Derigs: No conflict of interest disclosed. Ralf-Dieter Hofheinz: Financing of Scientific Research: Ja, Firmen Sanofi, Roche, Merck, Amgen, Bayer, medac; Expert Testimony: Ja, Firmen Sanofi, Roche, Merck, Amgen, medac. P214

Intravenous ferric carboxymaltose vs. oral iron substitution in patients with metastatic colorectal cancer (mCRC) and iron deficiency anemia: a randomized multicenter treatment optimization study (a study in progress report) zur Hausen G.1, Rötzer I.2, Reichart A.1, Pauligk C.1, Hunfeld K.-P.3, Hozaeel W.1, Quidde J.4, Hofheinz R.-D.5, Al-Batran S.-E.1 Krankenhaus Nordwest gGmbH, Institut für Klinisch-Onkologische Forschung, Frankfurt am Main, Germany, 2Krankenhaus Nordwest gGmbH, Klinik für Onkologie und Hämatologie, Frankfurt am Main, Germany, 3Krankenhaus Nordwest gGmbH, Labormedizin, Mikrobiologie und Krankenhaushygiene, Frankfurt am Main, Germany, 4Universitätsklinikum Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany, 5Universitätsmedizin Mannheim, III. Medizinische Klinik, Mannheim, Germany 1

Introduction: Iron deficiency has a high prevalence in colorectal cancer patients ranging at ca. 60%. About 70% of these patients suffer from iron deficiency anemia (IDA) which adds both physical and cognitive impediments to an already straining chemotherapy. Moreover, a chronic disease like cancer often results in a reduced availability of iron for the body. In clinical practice iron substitution is usually administered orally. Due to low resorption rates, frequent gastric side effects and thus poor patient compliance a parenteral substitution seems to be a better option in terms of efficacy. In the framework of a randomized multicenter clinical trial (‘FerInject’) a comparison of efficacy parameters of parenteral vs. oral iron substitution will now be conducted in order to identify the best treatment form for clinical practice in oncology. Furthermore detailed quality of life-data (QoL) will be collected in both treatment arms for effect comparison. Methods: Randomized explorative multicenter phase II trial: mCRC patients with IDA and a concomitant palliative chemotherapy will be randomly assigned to one of the treatment arms (parenteral vs. oral). Parenteral patients receive up to 1.000 mg ferric carboxymaltose intravenously per week for a maximum of 2 weeks. Oral patients take 200 mg iron per day for a total of 12 weeks. Course of therapy and QoL-data will be collected. Primary endpoint is the increase or normalization of hemoglobin. Secondary endpoints are fatigue; QoL; handgrip strength; number of allogenic blood transfusions;

62

Oncol Res Treat 2015;38(suppl 5):1–270

time until rise or normalization of hemoglobin; genesis of the IDA; number, dose and duration of therapy with recombinant erythropoietin; inflammatory parameters; influence of nutritional status on IDA and therapy success; tolerance and toxicity; dropout rate and overall survival. 64 patients shall be enrolled at currently 12 study sites. Data will be evaluated exploratively due to lacking reference data. The development of hemoglobin status in the treatment arms will serve as an indicator for a trend towards a better treatment option if a level of significance p = 0.2 (Fisher’s exact test) can be detected. Results: Recruitment started in 04/2015. Conclusions: FerInject is a trial in progress with high relevance for the clinical management of mCRC patients with IDA. Furthermore it will return additional information to both efficacy and QoL issues of these patients under a concomitant palliative chemotherapy. Disclosure: Gerrit zur Hausen: No conflict of interest disclosed. Salah-Eddin Al-Batran: Expert Testimony: Research grant (Vifor)

Posterdiskussion Lymphome aggressiv P215

Systemic relapses in primary CNS lymphoma (PCNSL) Korfel A.1, Fischer L.2, Hummel M.3, Lenze D.4, Hommel A.1, Weller M.5, Roth P.5, Kreher S.1 Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité – Universitätsmedizin, Berlin, Germany, 2Zentrum für Onkologie und Urologie, Rostock, Germany, 3Institut für Pathologie – Molekulare Diagnostik, Charité – Universitätsmedizin, Berlin, Germany, 4Institut für Pathologie CBF – Molekulare Diagnostik, Charité – Universitätsmedizin, Berlin, Germany, 5 Department of Neurology University Hospital Zurich, Zürich, Switzerland 1

Introduction: PCNSL is a rare lymphoma confined to the CNS at the time of diagnosis. A strong affinity to the CNS is typical for PCNSL with systemic relapse (SR) occurring very rarely. Methods: We retrospectively analysed two prospective collectives of immunocompetent adult PCNSL patients: 411 from a multicenter therapy study (G-PCNSL-SG-1; collective 1) and 51 treated for PCNSL at Charité Berlin from 1994 to 2007 (collective 2). Within the collective 1, patients with SR were compared to patients who did not develop an SR during the whole follow-up. When possible, tumor tissue from SR was compared to the primary CNS tumor, and clonality analysis was performed using PCR for immunoglobulin heavy/light chain (IgH/L-PCR, Biomed2 protocol). Results: SR was found in 24 of 411 patients in collective 1 (5.8%) and in 6 of 51 (11.7%) in collective 2 for a total of 30 SR patients. Median time to the first SR was 25.3 months (range 2–97). SR was found at first relapse in 26 patients; at the second relapse in 6 and at the third relapse in 4 patients for a total of 36 SR. The majority of SR were localised extranodally and was not accompanied by CNS relapse (30/36 = 83% each). Median survival from the first SR was 9.9 months (95% CI 3.0–16.9). No significant differences in initial characteristics were found between SR and non-SR patients within collective 1. The outcome of SR versus non-SR patients was similar: median overall survival from PCNSL diagnosis 30.6 months (95% CI 0–64) versus 34.4 months (95% CI 28.5–40.3; p = 0.960); median progression free survival from PCNSL diagnosis to first relapse 15.5 months (95% CI 0–31.6) versus 12 months (95% CI 8.9–15.1), respectively (p = 0.355). Clonality analysis of SR and corresponding primary CNS tumor was performed in 9 patients. Concordant clonality was detected in 6 (67%), discordant clonality in 3 (33%) patients. Conclusions: SR is rarely diagnosed in PCNSL when restaging does not routinely include search for systemic disease. Patients who develop a SR do not show differences at the time of PCNSL diagnosis as compared to those who do not develop it. The usually extranodal SR localisation underlines the particular tropism of PCNSL. The prognosis of SR patients is comparable to that of non-SR patients. Most of SR showed the same Ig rearrangement as the primary CNS tumor. However, discordant clonality in

Abstracts


CONTENTS AUTHOR INDEX

one third of analysed patients indicated the systemic lymphoma as second malignancy independent of PCNSL. Disclosure: No conflict of interest disclosed. P216

Cotargeting of PIM, PI3K and mTOR in mantle cell lymphoma Freysoldt B.1,2, Schnaiter A.1,2, Zimmermann Y.1,2, Hutter G.1,2, Hiddemann W.1,2, Dreyling M.1,2 Helmholtz Zentrum, München, Germany, 2Medizinische Klinik und Poliklinik III, Klinikum Großhadern, LMU, München, Germany 1

Introduction: Mantle cell lymphoma (MCL) comprises about 6% of all non-Hodgkin´s lymphoma with a median survival of 3–5 years. The proviral insertion in murine (PIM) lymphoma proteins are serine/threonine kinases which play an important role in cell survival and proliferation. They are overexpressed in multiple human cancers, including haematological malignancies. On the other hand, PI3K inhibition achieves high, but temporary responses in relapsed MCL. In this study we evaluated the efficiency and mode of action of a dual PIM/PI3K and a triple PIM/PI3K/ mTOR-Inhibitor in MCL cell lines. Methods: MCL cell lines (Granta 519, Jeko-1, Rec-1 and Mino) were exposed to a dual PIM kinase/PI3K inhibitor (IBL202) and triple PIM-kinase/PI3K/mTOR inhibitor (IBL301). Cell proliferation (trypan blue staining), apoptosis (Annexin V PE/7-AAD staining) and cell cycle (FACS) were investigated. Protein expression and phosphorylation status of downstream proteins (Akt, GSK-3β, 4EBP1) as well as markers of apoptosis (PARP, Caspase 9) were analysed after 1h, 4h, 8h and 24h. Results: Both IBL202 and IBL301 appear to be effective inhibitors, with the triple inhibitor IBL301 being superior to the dual inhibitor IBL202. IBL301 had a much higher impact on cell proliferation than IBL202 in all tested MCL cell lines, possibly due to its mTOR inhibiting activity. Both, treatment with IBL202 and IBL301, induced cell death, but cell death rate was almost twice as high in IBL301 treated MCL cell lines. The differential impact of the two inhibitors could be also confirmed based on considerably higher PARP and Caspase 9 cleavage after treatment with IBL301. Both inhibitors led to G1 arrest. In Jeko-1, Granta-519 and Mino dephosphorylation of Akt was detected after treatment with both agents, again with a less prominent effect under treatment with IBL202, supporting the mode of action of both inhibitors through the PI3K-AKT pathway. Accordingly, dephosphorylation of GSK-3β was observed after both agents in all MCL cell lines. The decrease of phosphorylated proteins occurred during the first hour of treatment, while subsequently phosphorylation increased again up to the level of controls. Conclusions: The triple inhibitor of PIM kinases, PI3K and mTOR shows impressive efficiency in MCL cell lines, superior to the dual inhibition of PIM kinase and PI3K. Cotargeting PIM kinases, PI3K and mTOR represents a promising novel approach in MCL.

DLBCL were classified as non-GCB or GCB subtype according to the Visco-Young immunohistochemistry algorithm and analyzed for mutations in MYD88, CD79A/B, and CARD11 by Sanger Sequencing and real time PCR. Statistical comparisons were made using Fisher’s exact test. As expected, significantly more non-GCB lymphomas carried mutations in MYD88, CD79A/B, and CARD11 than GCB lymphomas (19/48 vs. 4/31 cases, p < 0.02) when only lymphomas of primary nodal origin were considered. In extra nodal lymphomas no difference in mutation pattern was seen between GCB and non-GCB type (overall mutations: 18/40 vs. 9/23, n.s.). Whereas CARD 11 mutations were evenly distributed among all entities investigated (GCB and non-GCB, nodal and extranodal), CD79 mutations were not detected in primary nodal GCB lymphomas and MYD88 mutations only in one case. Overall, the distribution of mutations in all primary extra nodal lymphomas strongly resembled the mutation pattern in nodal non-GCB lymphomas and was significantly different from GCB (30/70 vs. 4/31, p < 0.005). Whereas primary nodal DLBCL consist of two molecularly different entities (ABC and GCB) these molecular differences do not exist in primary extra nodal lymphomas. Although it is technically possible to use the Visco-Young algorithm to subtype extra nodal DLBCL the resulting categories do not reflect molecular subtypes. Primary extra nodal DLBCL should be regarded as subtype/subtypes of its own with some features shared with non-GCB DLBCL. References: 1 Nature. 2000:503–511. 2 Proc Natl Acad Sci USA. 2008:13520–13525. Disclosure: No conflict of interest disclosed. P218

MiR-199a and miR-497 are associated with better overall survival due to increased chemosensitivity in aggressive non-Hodgkin´s lymphoma patients Troppan K.1, Wenzl K.1, Pichler M.2, Pursche B.1, Schwarzenbacher D.3, Feichtinger J.4, Thallinger G.5, Beham-Schmid C.6, Neumeister P.1, Deutsch A.1 Medical University Graz, Hematology, Graz, Austria, 2MD Anderson Cancer Center, Department of Experimental Therapeutics, Houston, United States, 3 Medical University Graz, Oncology, Graz, Austria, 4University of Technology, Institute for Genomics and Bioinformatics/Institute of Biochemistry, Graz, Austria, 5Unversity of Technology, Institute for Genomics and Bioinformatics/ Institute of Biochemistry, Graz, Austria, 6Medical University Graz, Pathology, Graz, Austria 1

Since ABC (activated B-cell type) and GCB (germinal center B-cell type) have been described as separate entities of DLBCL (diffuse large B-cell lymphoma)1,2 efforts have been underway to define these entities on a molecular basis. In ABC lymphoma activation of the NF-kB pathway was described as a key feature and this activation has been linked to mutations in the B-cell receptor pathway and MYD88 mutations. The defining features of GCBs are far less well understood. Less attention has been paid to the primary localization of the tumor and its effect on mutation patterns.

Background: Micro-RNAs (miRNAs) are short non-coding single-stranded RNA molecules regulating gene expression at the post-transcriptional level. MiRNAs are involved in cell development, differentiation, apoptosis, and proliferation. MiRNAs can either function as tumor suppressor genes or oncogenes in various important pathways. The expression of specific miRNAs has been identified to correlate with tumor prognosis. Methods: For miRNA expression analysis real-time PCR on 81 samples was performed, including 63 diffuse large B-cell lymphoma (DLBCL, 15 of germinal center B-cell like subtype, 17 non germinal center B-cell, 23 transformed, and 8 unclassified) and 18 controls, including 9 peripheral B-cells, 5 germinal-center B-cells (GC), 4 lymphadenitis samples, and 4 lymphoma cell lines (RI-1, SUDHL4, Karpas, U2932). Expression levels of a panel of 11 miRNAs that have been previously involved in other types of cancer (miR-15b_2, miR-16_1*, miR-16_2, miR-16_2*, miR-27a, miR-27a*, miR-98–1, miR-103a, miR-185, miR-199a, and miR-497) were measured and correlated with clinical data. Furthermore, cell lines, lacking miR-199a and miR-497 expression, were electroporated with the two respective miRNAs and treated with standard immunochemotherapy routinely used in patients with DLBCL, followed by functional analyses including cell count and apoptosis assays. Results: 7 miRNAs (miR-16_1*, miR-16_2*, miR-27a, miR-103, miR185, miR-199, and miR-497) were statistically significant up-regulated in DLBCL compared to normal GC. However, high expression of miR-497

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. P217

ABC, GCB, and extra nodal DLBCL: Three sides of a story or three stories with similar sides? Bohlen J.1, Hallas C.2, Preukschaß M.2, Tiemann M.2 Semmelweis Universität Medizinische Fakultät, Asklepios Campus Hamburg, Hamburg, Germany, 2Hämatopathologie Hamburg, Molekularpathologie, Hamburg, Germany 1

63


CONTENTS AUTHOR INDEX

or miR-199a was associated with better overall survival (p = 0.042 and p = 0.007). Overexpression of miR-199a and miR-497 led to a statistically significant decrease in viable cells in a dose-dependent fashion after exposure to rituximab and various chemotherapeutics relevant in multi-agent lymphoma therapy. Conclusion: Our data indicate that elevated miR-199a and miR-497 levels are associated with improved survival in aggressive lymphoma patients most likely by modifying drug sensitivity to immunochemotherapy. This functional impairment may serve as a potential novel therapeutic target in future treatment of patients with DLBCL. Disclosure: No conflict of interest disclosed. P219

Outcome of patients with transformed CLL and FL compared to patients with relapsed DLBCL – retrospective analysis of the Freiburg cohort Bach A.1,2, Greil C.2, Marks R.2 Charité – Universitätsmedizin Berlin, Radiologie, Berlin, Germany, Uniklinik Freiburg / Innere Medizin / Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany 1 2

Introduction: Patients with transformed chronic lymphatic leukaemia (CLL) have poor prognosis despite dose-intensified therapies. We determined outcome parameters for all patients transformed from CLL or follicular lymphoma (FL) into diffuse large B-cell lymphoma (DLBCL), using patients relapsed from DLBCL as comparison group. Methods: All patients with either FL or CLL and subsequent transformation into DLBCL or relapsed DLBCL (rDLBCL) who were treated in the University Hospital Freiburg between 2000 and 2011 were analysed. Overall survival (OS) after transformation/relapse and disease free survival (DFS) after first complete remission (CR) were evaluated. Results: 98 patients were identified, 16 (16.3%) with transformed FL (tFL), 14 (14.3%) with transformed CLL (tCLL) and 68 (69.4%) with rDLBCL. At the time of transformation/relapse, patients with tFL, tCLL and rDLBCL were treated with either chemotherapy alone (25%, 64%, 49%), autologous (autoSCT; 56%, 0%, 38%) or allogeneic transplantation (alloSCT; 13%, 29%, 3%). While tFL and rDLBCL patients experienced a CR in 56%/57% with this first line treatment, only 21% of tCLL patients achieved CR. After a median follow-up of 6.8 years, this inferior response was followed by a five-year OS (5y-OS) of only 14%, compared to 38% for tFL (p = 0.1912) and 27% for rDLBCL patients. Only 7% of all tCLL patients experienced longterm disease control, exclusively after alloSCT. In contrast, of all tFL patients in CR after initial therapy, to date 67% showed no signs of disease. A low/low-intermediate International Prognostic Index (IPI) at the time of transformation/relapse resulted in a significantly better median survival – especially for tCLL patients (32 months vs. 1.5 months for patients with high/high-intermediate IPI, p = 0.019). Regarding therapy, tCLL patients treated with alloSCT had improved median OS compared with those who received chemotherapy only (42 vs. 6 months, p = 0.1132). For tFL patients treatment with autoSCT resulted in a better median OS (24 vs. 6.5 months with chemotherapy; p = 0.3654). Conclusions: Overall survival of transformed low grade lymphoma is strongly influenced by the underlying disease with more favourable outcome for patients with FL compared to CLL. Dose intense therapy including autoSCT results in the best treatment outcome for patients with tFL or rDLBCL, while longterm disease control of transformed CLL patients can only be achieved with alloSCT. Disclosure: No conflict of interest disclosed.

64

Oncol Res Treat 2015;38(suppl 5):1–270

P220

Superantigen recognition is a specific hallmark of mantle cell lymphoma-derived B-cell receptors in a substantial proportion of patients Fichtner M.1, Spies E.1, Seismann H.1, Dreyling M.2, Binder M.1, Klapper W.3, Trepel M.1,4 Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 2Department of Medicine III, University Hospital Grosshadern, Munich, Germany, 3Institute of Pathology, Division of Hematopathology, University Medical Center Schleswig-Holstein, Kiel, Germany, 4Department of Hematology and Oncology, Augsburg Medical Center, Augsburg, Germany 1

Introduction: Mantle cell lymphoma (MCL) is a B cell Non-Hodgkin’s lymphoma with a poor prognosis. The success of agents targeting the B-cell receptor (BCR) signaling suggests a major role for this pathway in mantle cell lymphoma. However, it remains unclear what antigens trigger BCR activation in this disease. Methods: We used an unbiased PCR approach to amplify and sequence the BCR heavy and light chains of 24 randomly chosen MCL patients. The variable regions of the MCL BCRs were cloned into an IgG or Fab expression vector and expressed using Sf9 cells. In total, we produced eleven different recombinant monoclonal IgG antibodies and five Fab-fragments. With this set of immunoglobulins, we performed phage display library screenings, Western blottings, and immunofluorescence stainings as well as Protein A-ELISAs and Protein A-immunoprecipitations. Results: In line with previous research, the BCRs of our cohort were strongly biased, showing a preference for the usage of vH3-family genes (11/24). Further, all BCRs bearing the vH3–21 heavy chain were paired with vL3–19 lambda light chain (5/24). In addition, 5 of 24 samples used the vH434-family. Nearly all heavy chain sequences were not or only minimally mutated (≥98% germline identity). In the majority of cases, selections with random peptide phage libraries did not enrich peptides binding specifically to these MCL BCRs. Together, this suggested that classical high-affinity antigen recognition may not play a leading role in BCR function of MCL. Therefore, we explored the hypothesis that MCL BCRs might be activated by low-affinity, complementarity determining regions (CDR)-independent, superantigen-like or glycosylation-mediated interactions. While no BCR acquired novel glycosylation sites, all VH3 family expressing BCRs (11/24 cases) showed sequence features indicative for interaction with the superantigen protein A, which could be confirmed by Fab binding and immunoprecipitation studies. Furthermore all VH4–34 expressing BCRs (5/24 cases) exhibited motifs associated with anti-i/I reactivity. Conclusion: Our findings suggest that the BCR pathway may be activated by CDR-independent mechanisms in a substantial portion of patients with mantle cell lymphoma. Disclosure: No conflict of interest disclosed. P221

Comparison of the efficiencies of different PI3K-Inhibitors in matle cell lymphoma Hutter G.1,2, Zimmermann Y.1,2, Zoellner A.1,2, Irrgang P.1,2, Arnd J.1,2, Hiddemann W.1, Dreyling M.1,2 Universitätsklinikum München-Großhadern, Med III, München, Germany, Helmholtz Institute Munich, CCG Leukemia, Munich, Germany

1 2

Introduction: Mantle cell lymphoma (MCL), is a distinct lymphoma subtype with an aggressive clinical course and a median survival of 3–5 years. New emerging strategies include inhibitors of the B-cell receptorpathway which is constitutively activated in MCL and plays a critical role in tumor growth and survival. In the present study we investigated different PI3K-Inhibitors (IPI-145, CAL101, A66, TGX221 and Bay236) targeting different isoforms of PI3K in MCL. Methods: MCL cell lines (Z-138, Mino-1, Granta-519, Jeko-1, Rec-1, Maver-1) were exposed to different PI3K-Inhibitors (IPI-145, CAL101,

Abstracts


CONTENTS AUTHOR INDEX

A66, TGX221, Bay 236) with or without murine feeder layer (M210B4). The effect of drugs was evaluated by cell count (trypan-blue staining), cell metabolism (WST-assay), cell cycle (FACS) and apoptosis (Annexin V PE/7-AAD staining). Subsequently, combinations with other inhibitors of the PI3K/mTOR pathway were analysed. Western blot analyses were performed after exposure to the various inhibitors and were correlated to the sensitivity of cell lines. Finally, efficiency of drug combinations were confirmed in primary patient samples. Results: The PI3K inhibitor- Bay236 was most effective in MCL cell lines followed by IPI-145, A66, TGX221 and CAL101 suggesting a higher efficiency by inhibiting multiple isoforms of PI3K. In patient samples Bay 239 also revealed the highest cytotoxicity followed by IPI-145, CAL101, TGX221 and A66 (75,1%; 65,9%; 50,6% and 40,3%, respectively). Comparing different combinations of single PI3K-isoform inhibitors the combination of A66 and CAL101 was most efficient resulting in a 65,2% reduction of cell count. Nevertheless a combination of multiple isoform-specific PI3K inhibitors (A66, TGX-221, CAL101) led to an even higher reduction of cell proliferation (33,4–66,6% ) than targeting only single isoforms (0–21,6%) confirming the superiority of targeting multiple PI3K isoforms. The higher cytotoxic effect was accompanied by higher dephosphorylation of phAKT, phRictor and phRSK. Results of the effect of PI3K inhibitors on MCL in the presence of a murine feeder layer will be shown. Conclusion: Based on the comparative analysis of PI3K inhibitors inhibition of multiple isoforms of PI3K appears to be most efficient in MCL. Even more importantly, the murine feeder layer enhanced the effect of PI3K inhibitors indicating that the microenvironment plays a critical role for the mode of action of such inhibitors of the B-cell receptor pathway. Disclosure: Grit Hutter: No conflict of interest disclosed. Martin Dreyling: Advisory Role: Scientific advisor, Bayer. P222

High dose Cytarabine and Thiotepa plus G-CSF is an effective combined salvage and stem cell mobilization regimen in central nervous- and advanced nodal lymphoma Schäfer H.S.1, Schlosser T.1, Schorb E.1, Fritsch K.1, Finke J.1, Illerhaus G.2 Universitätsklinikum Freiburg, Department innere Medizin, Klinik für innere Medizin 1, Freiburg, Germany, 2Klinikum Stuttgart, Klinik für Hämatologie, Onkologie und Palliativmedizin, Stuttgart, Germany 1

Introduction: For aggressive B-cell lymphoma high-dose chemotherapy (HDC) followed by hematopoietic stem cell transplantation (HSCT) is a common treatment approach. Therefor used induction and conditioning regimens have been extensively analyzed for response rates, survival rates and toxicity. The impact and effectiveness of mobilization regimen are much less investigated. Methods: AraC (3 g/m2) was administered for 2 subsequent days in combination with thiotepa (40 mg/m2) on the 2nd day. Stem cells were mobilized with subcutaneous G-CSF. Stem cell apharesis was performed at Freiburg medical center according to GMP standard. Response and toxicity data were retrospectively assessed from mediacal record. Results: We retrospectively analyzed 73 patients with primary (n = 59), secondary (n = 9) central nervous non-Hodgkin lymphoma (PCNSL, SCNSL) and advanced nodal lymphoma (n = 5) (ANL) treated with Cytarabine and Thiotepa (AraC/TT) plus G-CSF chemotherapy as mobilization regimen for peripheral blood stem cell (PBSC) collection at our institution. For all patients, proceeding to high-dose therapy with autologous stem cell rescue was intended. Patients received 112 cycles of this regimen in total . Stem cell collection was performed after first or second cycle. The number of CD34 positive cells mobilized after AraC/TT was 18,55 × 10e6/kg cells (median; range 86,16 × 106 –1,05 × 106). Dose of G-CSF administered was 3360µg (median; range 1500µg-7200µg) in total. PBSC collection of more than 2 × 106/kg CD34 positive cells was successful in 68 (93,2%) patients, for 57 patients (78,1%) this could be done in a single collection. Toxicity of this regimen was mild. Adverse events were reported in 25 patients (34,7%)

Abstracts

including febrile neutropenia (n = 19) and prolonged cytopenia leading to harvest failure (n = 3). All events were manageable. Response to therapy (defined as SD, PR or CR) was seen in 60 (90,9%) patients, major response (PR, CR ) was seen in 30 (45,5%) of all patients. Conclusion: We conclude that AraC/TT plus G-CSF has high anti lymphoma activity in PCNSL, SCNSL and ANL respectively with acceptable toxicity and excellent PBSC mobilizing characteristics. Disclosure: No conflict of interest disclosed. P223

The B-cell receptor pathway is highly actived in canine DLBCL cell line CLBL-1 Zoellner A.-K.1, Ruetgen B.2, Hutter G.1,3, Zimmermann Y.1,3, Mohring M.4, Hiddemann W.3, Hirschberger J.5, Dreyling M.1,3 Klinikum der Universität München, Medizinische Klinik und Poliklinik III, München, Germany, 2University of Veterinary Medicine, Vienna, Austria, 3Clinical Cooperative Group Leukemia, Helmholtz Center Munich, München, Germany, 4 University Hospital Klinikum rechts der Isar, Nuclear Medicine, München, Germany, 5Centre for Clinical Veterinary Medicine, LMU, Clinic of Small Animal Medicine, München, Germany 1

Background: Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of canine lymphoma and shows significant similarities in its clinical and biological presentation to human Activated B-Cell (ABC) subtype of DLBCL. Aims: Here we demonstrate the expression of hallmark kinases of the B-cell receptor pathway in the canine DLBCL cellline CLBL-1. Further we investigated the treatment with B-cell receptor pathway interacting compounds such as BTK inhibitor Ibrutinib and PI3K inhibitor Idelalisib and substances lately known for their anti-lymphoid activity. Methods: Established canine DLBCL (CLBL-1) and T-cell lymphoma (CL-1) cell lines were cultivated under standard conditions in RPMI and exposed to ascending doses of Idelalisib, Ibrutinib, Temsirolimus, BX912, Ku-63764,Enzastaurin and Bortezomib. Cell proliferation was determined after 48 hours based on WST cell proliferation assay. Western blotting was performed after 24h. All experiments were performed at least in triplicates. Results: In Western blot analysis kinases hallmarking the B-cell receptorPI3K-AKT pathway (AKT, PDK, PI3K, mTOR) and their phosphorylated isoforms were detected in CLBL-1 cells. Untreated CLBL-1 cells expressed p42/44,p38, MEK, GSK alpha and GSK beta and their phosphorylated isoforms as well as the cyclins CDK2, CDK4, CDK7, CDK9 but no cyclin D1. Significantly, treatment with only 1,25nM Ibrutinib induced in WST analysis a growth reduction to 45%, with 1µM arresting growth thoroughly. The PI3K-delta inhibitor Idelalisib showed dose dependent effects: 0,6µM reduced cell growth to 41%, whereas 5µM reduced proliferation to 13%. The mTor inhibitor Temsirolimus showed high efficacy: 1,25nM Temsirolimus reduced cell proliferation to 38%, while the mtorc1/mtorc2 inhibitor Ku-63794 induced at the dose of 0,25µM a reduction to 49%. CLBL-1 was also sensitive towards other compounds with anti-lymphoid activity such as the PDK-1 inhibitor BX-912 (0,25µM; 20%), the PKC inhibitor Enzastaurin (1,25µM; 52%) and the proteasome inhibitor Bortezomib (5nM; 50%). Conclusion: The detected activated B-cell receptor pathway in CLBL-1 and the sensitivity towards small molecule inhibitors targeting this pathway indicates the similarity to human ABC DLBCL. This data strongly supports the relevance of canine DLBCL as model for its human counterpart. Disclosure: Anna-Katharina Zoellner: No conflict of interest disclosed. Martin Dreyling: Financing of Scientific Research: Bayer: Speakers Honoraria; Expert Testimony: Bayer: Support of IITs.

Oncol Res Treat 2015;38(suppl 5):1–270

65


CONTENTS AUTHOR INDEX

P224

Xenograft mouse model of primary and secondary CNS lymphoma Isbell L.K.1, Reinacher P.C.2, Klingner K.3, Doostkam S.4, Tschuch C.3, Schorb E.1, Fritsch K.1, Schaefer H.1, Illerhaus G.5, Schaefer H.-E.6, Duyster J.1, Schueler J.3, von Bubnoff N.1 Uniklinik Freiburg, Klinik für Innere Medizin I, Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 2Uniklinik Freiburg, Abteilung für Stereotaktische und Funktionelle Neurochirurgie, Freiburg, Germany, 3Oncotest GmbH, Freiburg, Germany, 4Uniklinik Freiburg, Institut für Neuropathologie, Freiburg, Germany, 5Klinikum Stuttgart, Klinik für Hämatologie, Onkologie und Palliativmedizin, Freiburg, Germany, 6Uniklinik Freiburg, Institut für Klinische Pathologie, Freiburg, Germany 1

Background: Extranodal diffuse large B-cell lymphoma confined to the central nervous system defines primary CNS lymphoma (PCNSL). In addition, CNS involvement can occur in systemic diffuse large B-cell lymphoma (secondary central nervous system lymphoma, SCNSL). Mechanisms that commit lymphoma cells to the CNS compartment are poorly understood. Despite intensive treatment, many patients suffer from relapse. The generation of functional and reproducible in vivo models is a prerequisite for a better understanding of cerebral lymphoma and for the development of improved therapeutic strategies. Methods: We established a central nervous system lymphoma (CNSL) xenograft mouse model. Human PCNSL and SCNSL stereotactic biopsies are suspended into a transportation medium and directly transplanted subcutaneously in recipient NSG/NOG mice. Subcutaneous tumor growth was monitored by caliper measurement and mice were sacrificed with a tumor size of 2000 mm³. One third of the tumor as well as spleen, liver and brain were analyzed by histology and immunohistochemistry. One third of the tumor was snap frozen for subsequent genetic analysis, and one third was serially transplanted into NSG/NOG recipient mice. Results: We observed growth of transplanted cells from PCNSL samples (n = 4) and SCNSL samples (n = 3) at the subcutaneous site of NSG/NOG mice. Histology and immunohistochemical analysis of the xenograft tumors showed the same morphology and immunophenotype as the primary human CNSL samples. Histopathologic analyses of spleen, liver, bone marrow and CNS samples of the tumor bearing mice are under investigation. Conclusion: This xenograft mouse model allows in vivo studies on the biology of these rare tumors. Since PCNSL research is hampered by the small size of biopsy specimens, the expansion of the tumor cells in a xenografts model without losing the PCNSL phenotype may overcome this limitation. In addition, this tumor model allows us to compare lymphomas with and without CNS tropism with the goal to identify molecules that are critical for CNS tropism and thus could be potential new targets for therapeutic approaches. Furthermore this mouse model might be used to investigate novel pharmacologic agents to treat primary and secondary CNS lymphomas. Disclosure: No conflict of interest disclosed. P225

The modification of acid ceramidase activity and ceramides level as an indicator of the drug resistance in patients by lymphoma Abovyan M.1, Sahakyan L.1, Saharyan A.2, Shaljyan A.2 Haematology Center aft. Prof.R.Yolyan, Yerevan, Armenia, 2Yerevan State Medical University, Yerevan, Armenia 1

Aim: The involvement of sphingolipids, and their metabolites, such as ceramides in cancerogenesis was demonstrated in solid tumors as well as in hematological malignancies. As is known, the appearance of multidrug resistant phenotype (MDR) – one of the major causes of failures in the treatment of malignant diseases. According to recent years’ data, ceramide signaling plays an important role in tumor progression and development of chemoresistance. The present study was to examine the modification of

66

Oncol Res Treat 2015;38(suppl 5):1–270

acid ceramidase (aCD) activity and ceramides level as an indicator of the drug resistance in patients by lymphoma. Methods: The study included 123 patients with B-cell nonHodgkin’s lymphoma (NHL), which were admitted to the Hematology Center of Armenia. After obtaining informed consent were evaluated clinical and hematological parameters of all patients. Patients´ blood was taken before and after the treatment. For the normal control was used blood from 31 healthy donors of Hematology center. Ceramide has been determined using high performance liquid chromatography (HPLC). The aCDase activity determination was conducted according to the fluorogenic methods (Bedia C. et al., 2010). Results: According to obtained data two of 105 newly diagnosed and untreated NHL patients (1.9%) and seven of 18 previously treated patients and drug-resistant (38.9%) had detectable high levels of ceramides and twofold increased activity of aCD. Due to failure of therapy, 7 patients died in the treatment first trimesters, the remaining 2 patients 6 and 8 months ago. Development of MDR in NHL is in part driven by the inherent genetic heterogeneity and instability of the tumor cells. Our results suggest that ceramides level and ceramidase activity is a potential pharmacologic target for the NHL treatment. The inhibition of ceramides expression or ceramidase activity might represent a novel strategy to sensitize B-cell NHL patients to chemotherapy. Conclusions: Sphingolipids metabolites can be considered promising therapeutic tools alone or in combination with other compounds, as well as valid targets in the attempt to lymphoma treatment and overcome drug resistance. Disclosure: No conflict of interest disclosed. P226

Evaluation of safety, tolerability and efficacy of Temsirolimus in patients with relapsed or refractory mantle cell lymphoma (rel/refr MCL) in routine clinical practice Krekeler G.1, Neuhof A.1, Dreyling M.2, Hess G.3, Kalanovic D.1 Pfizer Pharma GmbH, Berlin, Germany, 2Ludwig-Maximilians-Universität, München, Germany, 3Johannes-Gutenberg Universität, Mainz, Germany 1

Background: Temsirolimus (TEM), an mTOR-inhibitor, is approved in the EU for the treatment of patients (pts) with relapsed or refractory (rel/ refr) MCL. A pivotal study demonstrated significantly longer progression free survival with TEM (175 mg weekly for 3 weeks followed by 75 mg weekly) in rel/refr MCL pts compared to investigator´s choice therapy (4.8 mo vs 1.9 mo; P = 0.0009). Yet only limited data is available on TEM in an unselected patient population during clinical routine. To evaluate the safety profile and efficacy of TEM in this rare tumor entity, further collection of data in a post-approval prospective non-interventional trial is useful. Methods: A German multicenter registry for rel/refr MCL pts treated with TEM was started in Germany in Oct 2009 (NCT00700258) with regulatory and ethic committee´s approval. Objectives are the evaluation of the safety profile, tolerability and anti-tumor activity of TEM as well as the profile, comorbidities, characteristics and the sequence of systemic therapies. Results: From Oct 2012 to Feb 2015, 27 study sites recruited 48 pts. Baseline characteristics: 72.9% male; median age 74.0 yrs; ECOG PS 0 or 1 in 83.0%, ECOG PS 2 in 17.0% of the pts. According to MIPI score 23.9%, 30.4%, and 45.7% of the pts are classified as low, intermediate and high risk at the time of enrollment. Bone marrow is involved in 41.7% of the pts. Median time between diagnosis and start of treatment with TEM is 3.1 yrs (range 0.4–14.9). The median number of prior therapies is 2.0 (range 1–10) with 45.9% treated in ≥4th line. Most common adverse events (≥15%) are thrombopenia (43.8%), anemia (25.0%), leukopenia (18.8%), general physical health deterioration (18.8%) and diarrhoea (16.7%). Severe adverse events (drug related) are general, metabolic, psychiatric, skin, renal, gastrointestinal, respiratory and blood system disorders (in 1 pt each) and infections (in 3 pts). Preliminary efficacy analyses are available for 31 assessable pts and show an objective response in 9 pts (1CR and 8

Abstracts


CONTENTS AUTHOR INDEX

PR, 29.0%), a clinical benefit (CR, PR, MR and SD) in 17 pts (54.8%) and PD in 14 pts (45.2%). Median PFS is 4.2 months (range 3.3–5.7). Conclusions: The registry was started to evaluate the safety and efficacy of TEM in pts with rez/refr MCL in the routine clinical practice. In this here included patient collective with 45.7% high-risk pts, TEM shows a predictable, manageable tolerability profile. Efficacy remains comparable with phase III data. Disclosure: Gabriele Krekeler: Employment or Leadership Position: employee of Pfizer Pharma GmbH, Germany. Daniel Kalanovic: Employment or Leadership Position: employee of Pfizer Pharma GmbH, Germany. P227

Preliminary clinical experience on the efficacy and feasibility of a new combination regimen consisting of Pixantrone, Etoposide, and Bendamustine with or without the addition of Rituximab in patients with relapsed/refractory aggressive non-Hodgkin lymphomas Panny M.1, d’Amore F.2, Nösslinger T.1, Jørgensen J.2, Silkjaer T.2, Leppä S.3, Zintl P.4, Theocharous P.4, Keil F.1 Hanusch Krankenhaus, Abteilung für Hämatologie und Onkologie, Wien, Austria, 2University Hospital Aarhus, Department of Hematology and Nuclear Medicine, Aarhus, Denmark, 3University Central Hospital Cancer Center Helsinki, Helsinki, Finland, 4CTI Life Sciences Ltd., London, United Kingdom 1

Introduction: With currently available therapies, relapsed/refractory aggressive NHL after high-dose therapy or, in not transplant-eligible patients, after first-line chemotherapy represents an unmet clinical need. Methods: evaluating a combination chemotherapy based on pixantrone (Pix), a novel aza-anthracenedione approved by EMA in multiply relapsed/refractory aggressive NHL patients. Etoposide+bendamustine were selected based on prior combination with Pix, and efficacy in salvage regimens in relapsed/refractory aggressive NHL. Schedule: Pixantrone50 mg/m2 i.v. day1+8, Etoposide100 mg i.v. day1, Bendamustine90 mg i.v. day1 ± Rituximab375 mg/m2i.v. day1 (PREBEN/PEBEN). Each cycle was given at 3-weekly intervals for a maximum of 6 cycles. All patients were assessed for chemosensitivity with PET/CT after cycle 1 or 2. G-CSF support was administered according to guidelines. Results: 10 patients with relapsed/refractory aggressive NHL were treated according to the PREBEN/PEBEN-schedule and were evaluable in terms of presentation, feasibility and response. The histological diagnoses were distributed as follows: relapsed/refractory DLBCL (N = 6), transformed follicular lymphoma (tFL;N = 2), post-transplant lymphoproliferative disease of DLBCL type (PTLD-DLBCL;N = 1), peripheral T-cell lymphoma (PTCL-NOS;N = 1; without rituximab). Based on PET/CT assessment performed already after 1course of treatment, the PREBEN/PEBEN regimen showed metabolic CR in 4/10patients (DLBCL;N = 4) and good PR in 3patients (DLBCL;N = 1,tFL N = 1, PTCL-NOS = 1) corresponding to ORR = 70% (CR: 40%; PR: 30%). No response was seen in 3/10 pts (DLBCL N = 1, PTLD-DLBCL = 1, tFL = 1) (PD: 30%). In both CR and PR patients, a marked reduction of the lesions was already detectable after the 1course of treatment. RD was in the range 5–14+months. The regimen was feasible and most patients received it as out-patients. The most common grade 3–4toxicity was of hematological type. Grade3–4 infections were seen in 40% of these heavily pretreated patient, but they were manageable and could be successfully treated. Conclusion: The PREBEN/PEBEN schedule is feasible and profound responses early in the course of therapy are not uncommon. A phase 1/ phase 2(extension)study is in preparation. Disclosure: Michael Panny: No conflict of interest disclosed. Felix Keil: Financing of Scientific Research: CTI, Roche.

Abstracts

P228

Burkitt lymphoma treated with a rituximab and methotrexate based regimen (GMALL B-ALL/NHL 2002): A single-center retrospective analysis of efficacy, toxicity and experience with relapsed/refractory disease Cremer M.1, Schwarzbich M.-A.1, Schöning T.2, Lisenko K.1, Ho A.D.1, Witzens-Harig M.1 Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany, 2Pharmacy, University Hospital of Heidelberg, Heidelberg, Germany 1

Introduction: Immunochemotherapy with a rituximab and methotrexate based regimen according to the GMALL B-ALL/NHL 2002 protocol has become the standard treatment for Burkitt lymphoma in Germany. Treatment consists of six 6-day cycles with rituximab, high-dose methotrexate, high-dose cytosine arabinoside, cyclophosphamide, etoposide, ifosphamide, corticosteroids and intrathekal chemotherapy, with dose reductions for patients over 55 years of age. Two additional doses of rituximab are administered as maintenance. While this treatment is curative in the majority of patients, there is a paucity of data concerning treatment options for patients with relapsed/refractory disease after treatment with this protocol. Methods: We retrospectively analyzed data from 38 patients who were treated for newly diagnosed Burkitt lymphoma according to the GMALL B-ALL/NHL 2002 protocol at our institution between 2003 and 2013. The probabilities for overall survival and progression-free survival were calculated by the Kaplan-Meier method. Toxicities were retrospectively assessed and categorized according to the Common Terminology Criteria v3.0. We individually reviewed each case with r/r disease and present an overview of post-induction therapy and outcome in these patients. Results: Of the 38 patients included in the analysis, 36 received the full protocol as planned. The response rate (CR/PR) was 97,4%. OS at 1 and 3 years was 83,9% and 77,8% respectively, PFS at 1 and 3 years was 78,9% and 76,1% respectively. Toxicities were mainly hematologic, infectious and gastrointestinal. The nine patients who relapsed after induction therapy were treated with individual approaches adapted according to comorbidities, age, duration of remission, and site of relapse. Three patients achieved a second remission (CR/PR). Eight out of nine patients with r/r disease died, seven from progressive disease and one from complications of allogeneic hematopoietic stem cell transplantation, while one patient is alive but suffers from a third relapse at time of writing. Conclusions: Our findings concerning response rate, OS, PFS and toxicity are consistent with reports from the literature, confirming that the GMALL B-ALL/NHL 2002 protocol is efficient and feasible for treatment of Burkitt lymphoma. The analysis of patients with r/r disease shows the lack of effective salvage strategies and the dismal prognosis of this subgroup, emphasizing the need for improved upfront treatment strategies in this setting. Disclosure: No conflict of interest disclosed. P229

Treatment of high risk aggressive B cell lymphomas with DA EPOCH R- a single center retrospective analysis Panny M.1, Keil F.1, Möstl M.1, Kornberger T.1, Menschel E.1, Simanek R.1, Nösslinger T.1 Hanusch Krankenhaus, Abteilung für Hämatologie und Onkologie, Wien, Austria 1

Introduction: Promising results in patients suffering from high risk DLBCL, Burkitt`s lymphoma (BL), mediastinal gray-zone lymphoma (MGZL) and primary mediastinal B cell lymphoma (PMBCL) treated with DA-EPOCH R have been reported. In our center high risk DLBCL – defined as double-hit/double hit score 2 or high risk IPI – BL, MGZL and PMCL are treated with DA EPOCH R. Methods: Retrospective analysis of toxicity and efficacy in DA EPOCH R treated patients. So far 11 previously untreated patients – 8 male, 3 female-

Oncol Res Treat 2015;38(suppl 5):1–270

67


CONTENTS AUTHOR INDEX

with a median age of 60 a (32–75 a) have been treated with a total of 48 cycles of DA EPOCH R: 7 DLBCL, 1 MGZL, 1 PMBCL, 2BL. Results: Grade III/IV ANC occurred in 71% of all cycles, thrombocytopenia III/IV° in 17%, grade III anemia in 4% of 48 cycles. Dose escalation was possible in 10 cycles (21%) – in none of patients aged > 65a dose escalation was possible. Due to peripheral sensory neuropathy II-III° in 6 patients Vincristine had to be dose reduced in 31% of all cycles. Other CTCAE grade III non-hematopoietic toxicities were: peripheral motor neuropathy, mucositis, colitis, hyperglycemia – each in 1 patient. So far 7 patients finished treatment: 3 in CR, 1 in PR, 1 relapsed early, 1 had treatment failure, 1 patient had to be switched to a less toxic regimen due to repeated febrile neutropenia. Conclusion: Although limited data DA EPOCH R seems to be a feasible treatment with acceptable toxicity. Only in young rather fit patients dose escalation seems to be possible. Disclosure: Michael Panny: No conflict of interest disclosed. Thomas Nösslinger: Financing of Scientific Research: Roche, Gilead, Janssen. P230

Diffuse large B-cell lymphoma in a patient with acute renal failure Übner M.1, Spriewald B.1, Mackensen A.1 Universitätsklinikum Erlangen, Medizinische Klinik 5 – Hämatologie und Internistische Onkologie, Erlangen, Germany 1

Diffuse large B-cell lymphoma (DLBCL) is the most frequent non-Hodgkin lymphoma which shows extranodal involvement in 30% and involvement of the genitourinary tract in about 2–3% of all cases which might be associated with a worse prognosis compared to nodal lymphoma. Manifestation in kidney is extraordinary and the limited reports indicate a poor prognosis. We report about a 59 year old female patient who presented with acute renal failure. Except nausea and fatigue the patient complained of no further symptoms. Arterial hypertension, hyperthyroidism and a Struma were already known. Creatinine increased to 3.11 mg/dl. Since no cause for renal failure was found, a biopsy of the left kidney was performed leading to diagnosis of DLBCL. A FDG PET/CT showed enrichment of the tracer in both kidneys, skeletal system and retroperitoneal lymph nodes, which were almost normally configured. Bone marrow was free of lymphoma. Renal function and initially elevated LDH have improved after the first application of R-CHOP. Therapy was continued with five cycles R-CHOEP and two cycles HD-MTX. The clinical response was excellent and currently the patient is awaiting re-stagning. Disclosure: No conflict of interest disclosed.

Posterdiskussion MDS / Stammzellen P231

The erythroid regulator Erythroferrone (ERFE) is differentially regulated in CD71+ erythroprogenitor cells of patients with myelodysplastic syndromes (MDS) and is associated with prognosis Mossner M.1, Stöhr A.1, Nolte F.2, Jann J.C.1, Fey S.1, Nowak V.1, Obländer J.1, Pressler J.1, Baldus C.D.3, Schulze T.J.4, Neumann M.3, Hofmann W.-K.1, Nowak D.1 Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany, 2St. Hedwig-Krankenhaus, Berlin, Germany, 3Charité Campus Benjamin Franklin, Berlin, Germany, 4Institut für Transfusionsmedizin, Mannheim, Germany 1

Introduction: Myelodysplastic Syndromes (MDS) are characterized by disturbed iron homeostasis. Recently, Erythroferrone (ERFE) was discovered as a novel regulator of iron homeostasis, which is selectively produced by bone marrow (BM) erythroprogenitor cells during hematopoietic stress and EPO stimulation (Kautz et al., Nature Genetics 2014). We therefore sought to examine the role of ERFE expression in CD71+ erythroprogenitor cells derived from patients with MDS and secondary acute myeloid leukemia (sAML). Methods: CD71+ erythroprogenitor cells were immunomagnetically isolated from mononuclear BM cells of patients suffering from MDS (n = 86, IPSS-low/int-1-risk n = 69, IPSS-int-2/high-risk n = 17), sAML (n = 18) and age-matched healthy donors (n = 17). In addition to CD71+ cells, CD34+, CD61+, CD15+ selected BM as well as CD3+ selected peripheral blood (PB) cells were collected from three MDS patients as well as two healthy young and two healthy old donors. ERFE expression was quantified by quantitative real time PCR. Patient follow up (FU) data was available for n = 55 MDS and n = 13 sAML samples. Results: ERFE expression analysis in the above described hematopoietic cellular subfractions revealed almost exclusive expression of ERFE in CD71+ erythroprogenitor cells. ERFE expression profiles acquired in CD71 positive cells revealed a highly significant mean overexpression in MDS patients: IPSS-low/int-1-risk (fold change (FC)=4.1, p < 0.0001), IPSS-int-2/high-risk (FC = 4.6, p = 0.0003) and sAML (FC = 6.5, p < 0.0001) relative to age-matched healthy controls. However, a distinct fraction of patients displayed ERFE expression levels similar or even lower than those measured in healthy donors. Univariate analysis of these opposed groups revealed that low abundance of CD71+ ERFE transcripts was significantly associated with inferior overall survival (OS) in MDS patients (median survival 1.7 years vs. not reached, p = 0.0066) and also sAML (median survival 0.1 vs. 0.8 years, p = 0.031). Conclusion: The current observation of significantly inferior survival probability for MDS and sAML patients with low ERFE expression levels indicates a potentially important biologic and clinical relevance of this novel regulatory gene in the pathogenesis of MDS. Consequently, regulation of aberrant levels of the erythroid hormone ERFE in MDS erythroprogenitor cells could provide a promising target for novel therapeutic avenues that mechanistically address dysfunctional erythropoiesis in MDS. Disclosure: No conflict of interest disclosed.

68

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

P232

Telomere length is significantly shortened in patients with aplastic anemia compared to hypoplastic myelodysplastic syndrome Bouillon A.S.1, Ferreira M.S.1, Panse J.1, Reinecke P.2, Schemenau J.3, Haas R.3, Brümmendorf T.H.1, Germing U.3, Beier F.1 Uniklinik Aachen, Klinik für Hämatologie, Onkologie and Stammzelltransplantation, Aachen, Germany, 2Institut für Pathologie, Uniklinik, Düsseldorf, Germany, 3Klinik für Hämatologie, Onkologie und Klinische Immunologie, Universitätsklinikum, Düsseldorf, Germany 1

Introduction: The majority of cases of acquired aplastic anemia (AA) is characterized by an autoimmune attack against the hematopoietic stem cell compartment leading to pancytopenia and bone marrow (BM) failure. Thus, immunosuppressive therapy (IST) represents the standard treatment for AA. Hypoplastic myelodysplastic syndrome (hMDS) frequently mimics the clinical findings of AA and proper clinical discrimination of hMDS from AA sometimes remains difficult. The underlying pathogenesis of hMDS is not fully understood. However, based on the close clinical relationship of AA and hMDS, an immune-mediated mechanism similar to AA is propagated. Supporting this hypothesis, hMDS can respond at least partially to IST and on the other hand, AA can clonally evolve to hMDS. Telomere length (TL) shortens with every cell division and reflects the replicative potential of somatic cells. In contrast to AA, the role of TL for disease pathogenesis in hMDS remains unclear. In this study we aimed to retrospectively investigate TL as possible marker to distinguish hMDS from AA. Material and methods: TL measurement was carried out using confocal Q-FISH protocol as published previously. TL was retrospectively analyzed from BM biopsies at diagnosis of 12 patients with hMDS and 15 patients with AA treated in the University Hospital of Düsseldorf. TL analysis was performed in single-blinded fashion. Mean age was 45.2 years in AA patients and 65.2 years in patients with hMDS. 28 patients (range 18–80 years) with diagnosed M. Hodgkin without BM affection were used as controls for linear regression and calculation of age-adapted TL difference. Results: First, we analyzed whether TL in AA and hMDS still correlates with age. In contrast to controls (R2 = 0.16, p = 0.03), TL in AA (R2 = 0.07, p = 0.32) and hMDS (R2 = 0.17, p = 0.15) was not significantly correlated with age. Using the controls to adjust for age, age-adapted TL difference was significantly shortened both in patients with AA (median: -2.96 kb, range –4.21 to 0.26, p = 0.001) and patients with hMDS (median: -2.26, range –3.85 to –0.64, p = 0.005). In direct comparison, telomere shortening was more accelerated in patients with AA as compared to hMDS (p = 0.048). Conclusion: We provide first data on TL in patients with hMDS using confocal Q-FISH. We observe that age-adapted TL is significantly shorter in patients with AA compared to hMDS. Further data are needed to evaluate the role of TL in the evolution of AA and hMDS.

hort with 1475 pts with more than 4% medullary blasts (mb), not treated with IC or allogeneic stem cell transplantation (SCT). Results: Median age at IC was 59 (18–79) years. Treatment schedules were ICE 27%, TAD 26%, Idarubicine and Cytarabine (23%) and others (24%). WHO types at IC were: RAEB I 4.8%, RAEB II 17.5%, CMML II 0.7%, AML (20–29% mb) 24.1% and 52.9% already progressed into AML (>30% mb) (8 pts missing information). 32% patients received IC at the initial diagnosis, 68% after a progress to either higher risk MDS or AML. 67% of all pts with known remission status (n = 240) achieved complete remission (CR), 17% partial remission (PR), and 16% did not respond. 82% of pts induced at first diagnosis reached CR, whereas only 60% of pts induced at progression reached CR (p = 0.001). Age above 60 years and presence of auer rods were associated with lower CR rates (73 vs 59%, p = 0.02; 92% vs 63%, p < 0.001). Pts who received IC showed a survival benefit in comparison with the control group (14 vs 23ms, p < 0.001). 33% of pts received a consolidation chemotherapy and lived significantly longer than those without (32 vs 22ms, p = 0.001). 68 (23%) pts received allogeneic SCT 5 ms (0–99) after IC. Pts lived significantly longer after allogeneic SCT (64 vs 21ms, p < 0.001). After censoring pts who received SCT at time of SCT survival in the induction group was still better than in the control group (14 vs 21ms p < 0.001). In multivariate analysis, age >60 years and high/ very high risk karyotypes according to IPSS-R were the most important parameters associated with survival. When ignoring karyotypes auer rods, achievement of CR, as well as age >60 were most important. Discussion: Induction chemotherapy prolongs survival in higher risk MDS pts, with approximately 20% long term survivors (>60ms). Pts benefit from consolidation therapy and CR rates are higher in pts treated at first diagnoses. Poor risk karyotype, higher age, and non-remission are associated with poor prognosis. Whenever possible pts should receive allogenic transplantation (50% of pts are still alive after 60ms). Disclosure: No conflict of interest disclosed. P234

Establishment of a novel “short tandem repeat” based multiplex-PCR assay for accurate del(5q) quantification using genomic DNA Jann J.C.1, Mossner M.1, Nolte F.2, Fey S.1, Nowak V.1, Obländer J.1, Pressler J.1, Fabarius A.1, Haferlach C.3, Hofmann W.-K.1, Nowak D.1 Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany, 2St. Hedwig-Krankenhaus, Berlin, Germany, 3MLL Münchner Leukämie Labor, München, Germany 1

Introduction: Patients (pts) with high risk MDS can be treated with intensive chemotherapy (IC). The aim of our study was to identify patients´ characteristics that may have relevance for the probability of complete remission or survival after IC. Methods: We retrospectively analysed 299 pts diagnosed between 1988 and 2014 at our department, and treated with IC and compared this co-

Introduction: Cytogenetic lesions play a key role in diagnosis and prognostication of hematologic malignancies. Such aberrations can be readily assessed via metaphase cytogenetics (MC), FISH and microarrays. However, these methods often depend on the availability of viable cells, large amounts of DNA or are very expensive. For accurate quantification of chromosomal deletions in minute amounts of DNA specimen we present a novel PCR-based assay for interrogation of del(5q) aberrations in myelodysplastic syndromes (MDS) that is based on measuring allelic loss at heterozygous short tandem repeat (STR) loci. Methods: Genomic DNA was extracted from bone marrow (BM) cells from MDS patients with del(5q). 12 fluorochrome-labelled PCR amplicons covering STR loci between chromosome 5q21 and 5q31 were generated from 10 ng DNA in a single multiplex-PCR reaction. Amplicons were subjected to capillary electrophoresis for allele ratio quantification of heterozygous STR loci. Deviations from equal allelic ratios in heterozygous STR markers in the tumour samples were used to calculate the fraction of cells carrying del(5q). Results: Using our novel assay for quantification of del(5q) burden, we identified an average of 8 heterozygous 5q STR marker per individual in a total of n = 696 samples from n = 67 MDS patients and n = 136 healthy donors. Our analysis revealed strong inter-marker correlations (average standard deviation (SD) = 2.3%) and high reproducibility in duplicate measurements of n = 328 samples (SD = 0.86%). Moreover, highly concordant results were observed for serial dilution series (r² = 0.96) and

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. P233

Intensive chemotherapy and long term outcome in patients with myelodysplastic syndromes Zadrozny N.1, Schuler E.1, Strupp C.1, Hildebrandt B.2, Kündgen A.1, Kondakci M.1, Haas R.1, Kobbe G.1, Germing U.1 Universitätsklinikum Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany, 2Universitätsklinikum Düsseldorf, Klinik für Humangenetik und Anthropologie, Düsseldorf, Germany 1

69


CONTENTS AUTHOR INDEX

paired analyses between interphase-FISH and our assay (r² = 0.94). Finally, screening of patient BM samples during Lenalidomide treatment reliably confirmed molecular remission as confirmed by MC. Conclusion: With the establishment of a highly multiplexed PCR assay for interrogation of STRs in deleted chromosomal regions we provide a highly adaptable technique for quantification of genomic aberrations. As exemplarily shown for del(5q), our assay provides strong reproducibility and concordance with established techniques. Requiring only minute amounts of DNA, this tool is highly suitable for copy number quantification when only residual archival DNA, e.g fragmented DNA from formalin-fixed samples, and no cells for FISH analysis are available. In summary, our newly developed assay provides a sensitive tool for quantitative copy number analysis for any large scale chromosomal deletion. Disclosure: No conflict of interest disclosed. P235

Piaza: Non-interventional study on efficacy and safety of azacitidine (Vidaza®) in patients with myelodysplastic syndromes (MDS, Int-2 or high risk), AML (WHO 20–30% blasts), or CMML (10–29% bone marrow blasts without myeloproliferative disorder) Wehmeyer J.1, Zaiss M.2, Losem C.3, Schmitz S.4, Neidig C.5, Teichmann B.5, Harde J.5, Trarbach T.5 Hämatologisch-onkologische Gemeinschaftspraxis, Münster, Germany, 2Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany, 3Praxis für Onkologie u. Hämatologie, Neuss, Germany, 4Gemeinschaftspraxis für Hämatologie und Onkologie, Köln, Germany, 5iOMEDICO AG, Freiburg, Germany 1

Introduction: Myelodysplastic syndromes (MDS) represent one of the most frequent haematologic diseases of the elderly. Treatment with hypomethylating agents, such as azacitidine (AZA), has become the standard therapy for higher-risk MDS patients (pts) who are ineligible for stem cell transplantation. AZA is associated with improved outcomes in MDS, including delayed AML transformation and prolonged survival in pts with higher-risk MDS and is also approved for CMML and AML pts with 20–30% blasts. Methods: This prospective non-interventional study collected data on the routine treatment of MDS (Int-2 or high risk), AML or CMML with AZA. Treatment with AZA was documented for one year followed by a one year follow-up phase. Data on demographics, tumour characteristics, treatment duration and reasons for treatment discontinuation was collected. The main endpoints are PFS, ORR, and safety parameters. Results: A total of 149 pts were enrolled in 31 German sites. Pts were diagnosed with primary MDS (59.7%), AML/CMML (37.6%) or secondary MDS (2.7%). Mean age was 75.5 (SD: ±7.4) years. At inclusion most pts had an ECOG PS of 0 or 1 (73.8%). Median PFS was 10.9 months (95% CI = 8.7–12.8). Parameters found to be relevant to PFS in a multivariate Cox-model were ECOG PS at baseline (P = 0.0018; HR = 2.23) and previous RBC transfusion (P = 0.0362; HR = 1.57). Pts with ECOG 0 had a longer PFS than those with ECOG 1 (18.4 vs. 9.8 months). Further, pts without the requirement for RBC transfusion prior to treatment appeared to have a longer PFS than those who did receive transfusions (12.5 vs. 8.6 months). Median OS was 14.1 months (95% CI 12.3–17.2). The 2-year survival rate was 28.9%. ORR was 41.6% (CR: 10.7%; PR: 30.9% vs. 28.2% SD; 6% PD (remaining 24.2% missing data)). Median TTD was 6.8 months (95% CI 5.4–7.9). Most common AEs with suspected relation to AZA were gastrointestinal disorders (27.5%), such as nausea (20.8%) and vomiting (9.4%), blood and lymphatic disorders (23.5%) such as leukopenia (12.8%), anaemia (10.1%), thrombocytopenia (10.1%). Conclusions: AZA is an effective and well tolerated therapeutic option for pts with higher-risk MDS. ECOG PS and previous RBC transfusion were found to be a predictor of PFS. The response rate was higher than reported in pivotal trials. Median OS was 14.1 months, which is lower than reported in study AZA 001, the registration trial for AZA. This may be due to the fact that pts were 5 years older than in study AZA 001.

70

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: Jürgen Wehmeyer: Advisory Role: AMGEN, Bayer, BMS, Celgene, Roche; Stock Ownership: iOMEDICO AG; Financing of Scientific Research: AMGEN, Bayer, BMS, Celgene, Roche. Tanja Trarbach: Employment or Leadership Position: iOMEDICO AG. P236

Identification of somatic mutations by next-generation sequencing in secondary myelodysplastic syndromes of multiple myeloma patients Murga Penas E.M.1, Pomplun C.1, Paul U.1, Nagel I.1, Becher C.1, Gramatzki M.2, Günther A.2, Siebert R.1 Institute of Human Genetics, University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University Kiel, Kiel, Germany, 2University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University Kiel, Division of Stem Cell Transplantation and Immunotherapy, 2nd Medical Department, Kiel, Germany 1

Introduction: Life expectancy of patients with multiple myeloma (MM) has been improved with the advent of novel treatments based on high-dosage chemotherapy combined with autologous stem cell transplantation and novel agents (immunomodulatory drugs and proteasome inhibitors). However, improvement in survival has led to increasing risk of secondary malignancies in MM especially for the development of myelodysplastic syndrome (secondary sMDS). Methods: Three cases of MM patients diagnosed in addition with a sMDS were analyzed by karyotyping, fluorescence in situ hybridization (FISH), and next-generation sequencing (NGS). NGS was performed on DNA from CD138-negative cells of each patient using the MiSeq benchtop sequencer and the MiSeq Reporter and Illumina VariantStudio 2.2 for data analyses (Illumina, San Diego, USA). In case 1, a 49-years-old female (stage IIIA), and in case 3, a 55-years-old male (stage IIIA), had been treated with melphalan, cyclophosphamide, bendamustine, and lenalidomide. In case 2 a 60-years-old male (stage IIIA) had been treated with melphalan and cyclophosphamide but never had received an immunomodulatory drug. sMDS was diagnosed 7-, 8-, and 4-years after diagnosis of MM in case 1, 2, and 3, respectively. Results: FISH analyses on CD138-positive cells at time of diagnosis or relapse revealed a complex karyotype with a t(4;14)(p16:q32)/MMSET/ FGFR3-IGH in case 1 (non-hyperdiploid MM) and gains of chromosomes 5, 11, and 21 in cases 2 and 3 (hyperdiploid MM). At the time of sMDS diagnosis, standard cytogenetic analyses revealed a monosomy 7 (–7) in all cases. In the two hyperdiploid MM, –7 was part of complex karyotypes with additionally –5/5q- and –12. By NGS, case 1 showed a heterozygous c.408T>A mutation in the RUNX1 gene. Cases 2 and 3 showed both heterozygous mutations in splice sites of the TP53 gene, c.783–2A>G and c.375+2T>G, respectively. All detected mutations by NGS could be validated using Sanger sequencing. Conclusions: Here, evidence is provided that somatic mutations of RUNX1 and TP53, which are associated with advanced disease, occur recurrently in sMDS of MM patients. Moreover, we show –7 and –5/5q- to be recurrent chromosomal changes in sMDS in both hyperdiploid and non-hyperdiploid MM. Additional studies of a larger group of patients will be required to determine the incidence of these mutations in sMDS occurring in MM patients. This work was supported by the Medical Faculty of the Christian-AlbrechtsUniversity. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

P237

Is this scleroderma? Skin lesions in patient with profound cytopenia Ihne S.M.1, Karsten A.1, Schmidt M.1, Kerstan A.2, Einsele H.1, Grigoleit G.U.1, Knop S.1 Universitätsklinikum Würzburg, Medizinische Klinik II, Hämatologie, Würzburg, Germany, 2Universitätsklinikum Würzburg, Dermatologie, Würzburg, Germany 1

Accompanying autoimmune phenomena are observed in about 10% of patients with myelodysplastic syndrome (MDS). Manifestations may be unspecific, thereby impeding the diagnosis of the underlying disease. The pathomechanism behind is still rarely understood. Here we report a case of a 51-year-old man with severe fatigue, asthenia, weight loss (14 kg/10 months) and generalized scleroderma which started from the distal limbs and resulted in progressive limitation of motility. Noteworthy, those signs suggestive for connective tissue disease had been preceded by a seemingly idiopathic deep vein thrombosis. Concomitantly, we observed pancytopenia: leukocytes, 1.9 x 103/ul; neutrophils, 650/ul; eosinophils, 100/ul; hb, 10.2 g/dl; MCV, 99,3 fl; MCH, 34,3 pg; MCHC, 34,6 g/dl; platelets, 9 x 103/ul). Vitamin B12 deficiency was substituted without improvement of the haemogram, the level of folic acid was normal. Spleen was moderately enlarged. Skin biopsies showed generalized deep morphea with scattered figures of hemophagocytosis. Bone marrow aspirates revealed reactive changes without manifest signs of dysplasia or lymphoma infiltration. Erythropoiesis was increased with left shift, but megakaryocytes were nearly absent. Viral infections (Parvovirus B19, CMV, VZV, EBV, HHV6, HSV1/2, adenovirus) could be excluded. Astonishingly, augmented phagocytes with hemophagocytosis figures were found suspect for secondary hemophagocytic lymphohistiocytosis (HLH/MAS). According to the criteria for HLH, only 4 of 8 were fulfilled and mutation analysis of perforin showed a wild type gene, making an HLH unlikely. Because of rapidly progressing skin lesions, we decided on an immunosuppression, but neither high-dose steroids nor application of cyclophosphamide improved pancytopenia or generalized morphea. In view of increasing need of transfusion and progressively increased MCV another bone marrow biopsy was performed, then showing unequivocal signs of dysplasia of erythropoiesis and granulopoiesis. As synopsis of our findings, we interpreted the deep morphea as a paraneoplastic event preceding overt MDS. Because of the young age, increasing transfusion frequency and rapid progressive scleroderma with resulting limitations of respiratory movements, the patient underwent allogeneic stem cell transplantation. In the course of treatment skin lesions improved rapidly. Restaging 30 days after allogeneic SCT showed reactive changes without signs of dysplasia. Disclosure: No conflict of interest disclosed. P238

Optic disc swelling and preretinal Hemorrhage in Patient with Myeloblastic Syndrome Kochkorov A.1, Bauer G.1, Killer H.1 Kantonsspital Aarau AG, Augenklinik, Aarau, Switzerland

1

Background: The term Myelodysplastic Syndrome (MDS) is a set of mostly acquired, rare congenital, clonal diseases of the hematopoietic stem cells with heterogeneous clinical presentation. History and Signs: A 16-year-old patient was presented due to a gray spot in front of his left eye. Otherwise, he had no complaints, especially no dizziness and no fever. There was an optic disc swelling of both eyes, a preretinal haemorrhage in front of the macula with reduced visual acuity (0.2) of the left eye and pancytopenia. Therapy and Outcome: The patient was assigned into the children´s hospital for further clarification. Until the diagnosis was confirmed he got erythrocyte- and platelet- transfusions. Under this therapy his visual acuity on the left eye begun to improve and the funduscopic abnormalities started to reduce. After the refractory cytopenia type of MDS was

Abstracts

confirmed, a stem cell transplant was performed. From that time on and for more than 1 year, he has normal visual acuity and completely normal funduscopic findings. Conclusions: For unexplained retinal hemorrhages and optic disc swelling an underlying hematological disease should be excluded. Usually in MDS with ocular involvement is only the treatment of the underlying disease sufficient. Disclosure: No conflict of interest disclosed. P239

Additional plerixafor to granulocyte colony-stimulating factors for haematopoietic stem cell mobilisation for malignant lymphoma or multiple myeloma patients: A systematic review with meta-analysis Hartmann T.1, Hübel K.2, Monsef I.1, Engert A.2, Skoetz N.1 Uniklinik Köln, Evidenz-basierte Onkologie, Klinik I für Innere Medizin, Köln, Germany, 2Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany 1

Introduction: Autologous stem cell transplantation is widely used to restore functioning bone marrow in malignant lymphoma or multiple myeloma patients after myeloablative chemotherapy. Results of clinical trials indicate that plerixafor plus G-CSF leads to an increased mobilisation and release of CD34+ cells, facilitating effective apheresis. To evaluate the efficacy and safety of plerixafor we conducted a systematic review and meta-analysis. Methods: We searched Cochrane Central Register of Controlled Trials and MEDLINE (1990 to April 2015) for RCTs and included trials evaluating additional plerixafor compared to placebo or no therapy for stem cell mobilisation. Two review authors independently screened the results of the search strategies extracted data, assessed quality and analysed data. Final interpretation was done together with an experienced clinician. Results: We identified four trials fitting the inclusion criteria. However, one was closed prematurely and did not report results, another was not published at all (completed in 2009). The remaining two trials evaluated 600 participants with multiple myeloma or Non-Hodgkin lymphoma. No evidence for differences between plerixafor and control group regarding mortality at 12 months (RR 1.00; 95% CI 0.59 to 1.69; P = 1.00) and adverse events in study period one emerged in the meta-analysis. The meta-analysis showed a significant advantage for those patients randomised to plerixafor for achieving a targeted stem cell number in four or less apheresis days (RR 2.42, 95% CI 1.98 to 2.96; P < 0.00001). As there is high heterogeneity between studies for the number of transplanted patients, we did not meta-analyse these data. In AMD3100–3102MM 95.9% in the plerixafor arm and 88.3% in the placebo arm underwent transplantation (RR 1.09, 95% CI 1.02 to 1.16), in AMD3100–3101NHL 90% versus 55.4% (RR 1.62, 95% CI 1.39 to 1.89). None of the trials reported quality of life. Conclusion: The two trials, both conducted by Genzyme Corporation, the manufacturer of plerixafor, were published several times. Two other trials are not published at all. Therefore the meta-analysis must be interpreted with caution as there is a high risk for publication bias. There are hints for an increased stem cell collection in a shorter time, without affecting overall survival or adverse events. For a better comparability of GCS-F plus plerixafor and GCS-F alone, all conducted RCTs must be published. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

71


CONTENTS AUTHOR INDEX

P240

Quantification of the time and effort associated with autologous peripheral blood stem cell mobilisation: A European perspective Hübel K.1, Ostermann H.2, Noppeney R.3, Glaß B.4, Berger K.2, Reitan J.5, Mohty M.6 Universität Köln, Klinik I für Innere Medizin, Köln, Germany, Universitätsklinikum, München, Germany, 3Universitätsklinik, Essen, Germany, 4 Asklepios Klinik St. Georg, Hamburg, Germany, 5RJM Group, Crown Point, United States, 6Saint-Antoine Hospital, Paris, France 1 2

Introduction: Plerixafor is indicated in combination with G-CSF to enhance mobilisation of hematopoietic stem cells to the peripheral blood (PBSC) for collection. The aim of this non-interventional study was to assess resource utilization, including time, effort and costs to the hospital, associated with PBSC mobilisation and apheresis. Patients and methods: The study population includes patients with a primary diagnosis of NHL who underwent PBSC mobilisation at European centres. Part I of the study, currently ongoing, is a retrospective medical record review of 200 NHL patients from 9 centers across France and Germany. Selected patients are evenly divided between two eras: 1) prior to approval of plerixafor (until 1 June, 2009), pre-P era, and 2) after approval of plerixafor (1 July, 2010 and onwards), P era. Outcome measures include number of visits for administration of mobilising agents; duration (days) of administration of mobilising agents; agents used as mobilising agents; adverse events (AEs); number and hours of apheresis sessions; attainment of CD34+ target (yes, no) and days until CD 34+ target level was met. Part II of the analysis is an ongoing prospective time and motion evaluation of apheresis performed at each center. Time-motion assessments will be obtained retrospectively (Part I) and prospectively (Part II). Costs will be evaluated and quantified through micro-costing group interviews with local hospital administration.The primary study end point is difference in mean time/effort to perform apheresis (including apheresis related AEs, if any) and total costs associated with mobilisation to the hospital between patients in the “Pre-P” versus “P eras”. Results: At time of abstract submission, data collection is ongoing at all centers. Final results from four German centers will be presented. Comparisons will be made between the German centers and the overall study results. It is hypothesized that the key findings of this study will demonstrate the favorable impact of novel Interventions on the number of apheresis procedures required to reach a target PBSC, and failure rate of mobilisation, thus translating into reduced total transplant costs without increasing toxicity. Conclusion: The financial implications for transplant centers could be significant and may lead to further studies aiming to optimize staff time and resource utilization related to apheresis in real-world practice. Disclosure: Kai Hübel: Advisory Role: Beratung Sanofi; Financing of Scientific Research: Vortragshonorar Sanofi; Expert Testimony: Sanofi; Other Financial Relationships: Reisekostenerstattung Sanofi. Mohamad Mohty: Advisory Role: Beratung Sanofi; Financing of Scientific Research: Vortragshonorar Sanofi; Expert Testimony: Sanofi; Other Financial Relationships: Reisekostenerstattung Sanofi. P241

Addition of plerixafor to stem cell mobilization regimens in autologous donors with multiple myeloma and lymphoma can increase the efficacy of stem cell harvesting in poor mobilizers Stümpel J.-P.1, Jentzsch M.1, Ruschpler E.1, Leiblein S.1, Franke G.-N.1, Schwind S.1, Jäkel N.1, Wang S.-Y.1, Heyn S.1, Pönisch W.1, Niederwieser D.1, Vucinic V.1 Universitätsklinikum Leipzig, Hämatologie und Internistische Onkologie, Leipzig, Germany 1

Introduction: Plerixafor is a potent CXCR4 receptor antagonist that is licensed for stem cell mobilization in autologous stem cell donors with

72

Oncol Res Treat 2015;38(suppl 5):1–270

multiple myeloma or lymphoma who show poor mobilization after having been stimulated with recombinant granulocyte colony stimulating factor (r-metHuG-CSF, Filgastrim). Patients and methods: We performed 27 stem cell collections in 21 poor mobilizing autologous donors with multiple myeloma (n = 13) or malignant lymphoma (n = 8) adding plerixafor to the stimulation regimen with recombinant G-CSF from January 2011 till April 2014. At stem cell collection, our patients (9 males, 12 females) had a median age of 61.5 years (y) (range 44–72y). In all cases a large volume leucapheresis (maximal 4 × total blood volume) was performed. Patients received 20 µg/kg body weight (BW) plerixafor after having achieved peripheral reconstitution of white blood cells (WBC, >1 Gpt/l). 6 patients received a second dosage of plerixafor due to insufficient CD34+ cell harvesting. Results: The median concentration of CD34+ cells in peripheral blood (PB) before and after the first application of plerixafor was 7.8/ µl (range 2.9–14.3 /µl) and 21.2 /µl (range 10.2–54.1 /µl) respectively, resulting in a median 3.3-fold increase (range 1.4–5.3-fold). The median CD 34+ yield for all patients was 3.7 × 10^6/kg BW (range 1.6–12.8 × 10^6/kg BW) with median WBC counts of 857.3 × 10^8 (range 446–2214 ×10^8) in the product. The vitality of the cells after thawing was tested with methan blue and showed excellent in all cases (median 86.9%, range 73.3–92%, standard deviation 4.0%). Until today, 18 patients received the autologous stem cell transplantation. All of them showed good peripheral WBC recovery, in median on day 11 (range 10–15). Discussion: Our data shows that addition of plerixafor increases the efficacy of stem cell harvesting in poor mobilizers. The produced stem cells fulfilled the required quality control criteria like vitality and building of colony forming units (CFU). We identified a median 3.3 fold increase of CD34+ concentration in peripheral blood after the application of plerixafor in poor mobilizers. Thus, the mobilization outcome of our patients receiving plerixafor shows acceptable efficacy within the expected range. Disclosure: No conflict of interest disclosed. P242

A panel of mass spectrometry based serum protein tests for predicting Graft-versus-Host Disease (GvHD) and its severity Koldehoff M.1, Roder J.2, Hoffmann A.C.3, Roder H.2 University of Duisburg-Essen, Department of Bone Marrow Transplantation, Essen, Germany, 2Biodesix, Inc. Steamboat Springs, Colorado, United States, 3 University of Duisburg-Essen, Department of Medical Oncology, Essen, Germany 1

Introduction: Transplants of pluripotent hematopoietic stem cells (PHSC) or bone marrow (BM) are effective therapies against hematological malignancies. However, GvHD remains a major source of morbidity and mortality post-transplant. Identification of patients likely to develop severe forms of acute or chronic GvHD could allow the selection of more aggressive therapeutic regimens for patients assessed to be at high risk. Methods: Serum samples and clinical data were available from 124 patients (age 18–70) who had received PHSC or BM transplants. Five patients suffered no GvHD, 15 de novo chronic GvHD (cGvHD), 21 acute GvHD (aGvHD) but no cGvHD and 83 both aGvHD and cGvHD. Of patients with aGvHD 51% had grade I disease and of patients with cGvHD 53% had limited disease. Matrix assisted laser desorption/ionization (MALDI) mass spectra were acquired from the samples using the deep MALDI method, allowing a deep probing of the proteome. The spectra were preprocessed and spectral features defined. The integrated intensities of these features were combined with the clinical data using deep learning based machine learning techniques to create classifiers able to stratify patients into groups depending on occurrence and severity of GvHD. Results: Classifiers could be developed with significant power to predict occurrence and severity of GvHD. The area under the curves (AUCs) obtained for the clinical questions investigated and examples of the sensitivity and specificity achievable are summarized in the table (Occurrence of GvHD).

Abstracts


CONTENTS AUTHOR INDEX

P244

Tab. 1. Occurrence of GvHD.

AUC

Sensitivity/Sensitivity

aGvHD ?

0.74

63%/80% or 80%/65%

Grade II or worse aGvHD?

0.65

60%/67%

cGvHD post aGvHD?

0.75

63%/75%

Extensive vs limited cGvHD?

0.68

67%/69%

Conclusions: It is possible to provide information on occurrence and severity of GvHD from mass spectral analysis of post-transplant serum samples. If validated, this panel of tests could provide additional information useful for clinicians choosing treatment regimens for patients following PHSC or BM transplants. Disclosure: Michael Koldehoff: No conflict of interest disclosed. Heinrich Roder: Employment or Leadership Position: Biodesix, Inc. Steamboat Springs, CO, USA. P243

Mobilization and hematopoietic stem cell collection in patients with autoimmune disease Lisenko K.1, Pavel P.2, Ho A.D.1, Wuchter P.1, Blank N.1 Universitaetsklinikum Heidelberg, Innere Medizin 5, Heidelberg, Germany, 2Universitaetsklinikum Heidelberg, IKTZ Institut für Klinische Transfusionsmedizin und Zelltherapie, Heidelberg, Germany 1

Introduction: High-dose chemotherapy followed by autologous hematopoietic stem cell (HSC) transplantation is a promising therapeutic approach in patients with refractory systemic sclerosis (SSc), multiple sclerosis (MS) and other autoimmune diseases (AID). We investigated the feasibility and efficacy of HSC mobilization in AID-patients. Methods: This is a single center retrospective analysis of HSC mobilization and collection in 33 patients with refractory SSc (n = 15), MS (n = 11) and other AID like vasculitis, connective tissue disease or rheumatoid arthritis (n = 7). HSC mobilization with cyclophosphamide + G-CSF and HSC collection was performed between 1999 and 2015 at our institution. The impact of age, body weight, diagnosis, disease duration, skin sclerosis and previous cyclophosphamide treatment upon HSC collection parameters was investigated. The efficacy of two different mobilization regimens (cyclophosphamide 2 × 2 g/m2 versus 2 × 1 g/m2) was analyzed. Results: HSC collection was successful in all 33 patients. The median number of collected HSCs was 12.2, 8.0 and 8.2 CD34+ cells/kg ×106in SSc, MS and other AID. Twenty-five of 33 (76%) patients achieved a sufficient collection during one leukapheresis session, while 5 of 33 (15%) required two and 3 of 33 (9%) required three or more leukapheresis sessions. No correlation of the collected HSC number was observed regarding age, body weight, diagnosis, disease duration, skin sclerosis or previous cyclophosphamide treatment. Mobilization chemotherapy with cyclophosphamide 2 × 2 g/m2 (n = 16), 2 × 1 g/m2 (n = 15) or 2 × 0.5 g/ m2 (n = 1) resulted in equally high numbers of collected HSCs by leukapheresis on day 13 or 14. One patient collected HSC without mobilization chemotherapy. Conclusions: We could demonstrate that HSC collection is safe and feasible in patients with AID irrespective of age, body weight, diagnosis, disease duration and previous cycles of therapy. Mobilization chemotherapy with cyclophosphamide 2 × 1 g/m2 or 2 × 2 g/m2 and HSC sampling efficacy is equally effective in those patients. Disclosure: No conflict of interest disclosed.

Donor evaluation before syngeneic hematopoietic stem cell transplantation for high-risk B-cell chronic lymphocytic leukemia Klink A.1, Eigendorff E.1, Schmidt V.1, Dornaus S.1, Hilgendorf I.1, Hochhaus A.1, Sayer H.G.1,2 Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Onkologie, Jena, Germany, 2HELIOS Klinikum Erfurt, 4. Medizinische Klinik, Hämatologie und internistische Onkologie, Hämostaseologie, Erfurt, Germany 1

Introduction: Allogeneic hematopoietic stem cell transplantation (HSCT) is an established treatment in high-risk B-cell chronic lymphocytic leukemia (B-CLL). Lower relapse rate in lymphoma patients is observed after allogeneic including syngeneic transplantation when compared with autologous transplantation. Few data for syngeneic HSCT in B-CLL show promising results regarding long-term survival. The advantages are the absence of graft rejection and graft versus host disease. Epidemiological data confirmed significant higher disease risk among 1st degree relatives. Patient and methods: We report on a female patient, who was diagnosed with B-CLL, stage Binet A, in 9/2003. In 2010 the patient required treatment due to progressive leucocytosis and lymphadenopathy. Former treatment with chlorambucil/prednisolone resulted in stable disease parameters. Because of significant disease progression into Binet C with evidence of IGH gene aberration and a deletion (17p)/TP53 mutation in 03/2012 second-line treatment with five courses of Rituximab (R) and fludarabine/cyclophosphamide was initiated. In the presence of poor-prognostic marker and partial remission of CLL the patient was transferred to our center in order to evaluate allogeneic HSCT. The HSCT-comorbidity score revealed 3 points (infection [Lyme disease], asthma bronchiale) for the 64-year-old patient. Syngeneic transplantation was favored instead of matched unrelated HSCT. Results: Immunocytology of the twin sisters bone marrow excluded a monoclonal cell population since no cytogenetic evidence was found for the presence of malignant hematological disease, particularly B-CLL. The patient received 1.7 × 106 CD34+cells/kg after conditioning with R-BEAM from her identical twin sister in 09/2013. Early transplantation complications were stomatitis °I on day +3 and CMV-reactivation on day +15. Nineteen months after HSCT, the patient is still in complete remission of B-CLL with a Karnofsky index of 90–100%. The donor remains without any signs of a hematological disorder. Conclusion: Our case shows a favorable outcome for syngeneic HSCT for advanced high-risk B-CLL. To achieve long-term disease control a careful assessment of genetically identical twins regarding leukemic donor cell clone is advised. Disclosure: No conflict of interest disclosed.

Posterdiskussion Supportive Therapie P245

Efficacy of antibiotic prophylaxis in AML patients treated with low-intensity therapeutic regimens Bainschab A.1, Quehenberger F.2, Greinix H.T.1, Krause R.3, Wölfler A.1, Sill H.1, Zebisch A.1 Medical University Graz, Division of Hematology, Graz, Austria, 2Medical University Graz, Institute of Medical Informatics, Statistics and Documentation, Graz, Austria, 3Medical University Graz, Section of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Graz, Austria 1

Introduction: Survival of older patients with acute myeloid leukemia (AML) has significantly improved with the advent of low-intensity therapeutic regimens (LITR, comprising decitabine, azacitidine, and low-dose cytarabine). Yet, infectious complications are common during LITR treatment and might hamper the beneficial effect of these drugs. Therefore, we aimed to evaluate the incidence of and predisposing risk factors for infec-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

73


CONTENTS AUTHOR INDEX

tious complications, as well as the value of antibiotic prophylaxis during LITR treatment of AML. Methods: We retrospectively analyzed 39 AML patients, who were treated with 215 cycles of LITR at the Medical University Graz. LITR was administered first-line in 29 and after failure of conventional high-dose approaches in ten cases. Infection was defined as any event necessitating appropriate diagnostic measures and anti-infective treatment. Variables tested as risk factors were assessed at the beginning of each LITR cycle and comprised hemoglobin and transfusion dependence, neutrophil and platelet counts, lactate dehydrogenase (LDH), c-reactive protein (CRP), creatinine and glomerular filtration rate, precedent high-dose chemotherapy as well as antibiotic prophylaxis (fluoroquinolones in most cases). Risk factors for the occurrence of infectious complications were assessed by multivariate logistic regression. Results: Infectious complications occurred in 29/39 patients (74%) resulting in death in six cases (15%). Of the 215 LITR cycles administered, 53 (25%) were complicated by infections. A clinically diagnosed infection was reported in 27/53 cases (51%) with pneumonia being the predominant event. A microbiologically diagnosed infection was reported in 17/53 cases (32%), most frequently gram-negative bacteria. In a multivariate analysis of parameters assessed at the start of each LITR cycle, low hemoglobin (P = 0.009) and transfusion dependence (P = 0.001), as well as increased LDH (P = 0.006) and CRP (P = 0.048) levels independently predicted the occurrence of infection. Most importantly, antibiotic prophylaxis was associated to a decreased rate of infections (P = 0.010). Conclusions: Low hemoglobin and transfusion dependence, as well as high levels of LDH and CRP are valuable markers to predict infection during LITR treatment in AML. Additionally, antibiotic prophylaxis might help to reduce the incidence of infectious complications. These data suggest evaluation of antibiotic prophylaxis in this setting within a prospective clinical trial. Disclosure: Antonia Bainschab: No conflict of interest disclosed. Armin Zebisch: Financing of Scientific Research: Celgene. P246

Retrospective evaluation of the safety and efficacy of therapy with Vancomycin versus Linezolid in patients with febrile neutropenia following myelosupressive chemotherapy for hematologic malignancies in a real life setting Hegge N.1, Hahn-Ast C.2, Brägelmann J.2, Schwab K.2, Molitor E.2, Brossart P.2, Wolf D.2, Mayer K.2 Universitätsklinik, Bonn, Germany, 2Universitätsklinik Bonn, Medizinische Klinik 3, Bonn, Germany 1

Introduction: In patients with hematological malignancies and febrile neutropenia as a consequence of previous chemotherapy, broad-spectrum antibiotics are considered 1st line treatment. If fever persists and gram-positive infection is suspected Vancomycin (VAN) or Linezolid (LIN) represent potential options for escalation of antibiotic therapy. This single center retrospective analysis compared the efficacy and safety of adding VAN or LIN to an existing broad-spectrum antibiotic therapy. Methods: All patients with febrile neutropenia and normal renal function, in which VAN or LIN were part of the antibiotic escalation regimen treated in a tertiary care university hospital between 2010–2014 were identified and analyzed retrospectively. Results: 87 cases were identified (VAN: n = 62, LIN: n = 25). Baseline characteristics (gender, malignancy, comorbidity, age, neutropenia-duration, type of chemotherapy and renal function) were comparable between the groups. Median age was 56 years (46–62). No significant difference was observed in distribution of gram (+), gram (–) and mixed gram (±) pathogens. VAN or LIN was used as second escalation in the antibiotic regimen in more than 70% of patients. Because of persisting fever, antibiotic strategy had to be modified more often in the VAN-group as compared to the LIN-group (VAN 51.6%, LIN 24.0%, p = 0.03). The median days (d) with persisting fever after escalation to VAN or LIN was slightly shorter in the LIN-group (VAN = 3d, LIN = 2d, p = 0.13). 92.2%

74

Oncol Res Treat 2015;38(suppl 5):1–270

of all detected gram(+)-pathogens were glycopeptide-sensitive. Regarding toxicities there was no significant difference in renal function (glomerular filtration rate/GFR) during therapy (VAN 78.35 ml/min, LIN 90.20 ml/ min, p = 0.8). After escalation to VAN/LIN the LIN-group showed a trend towards shorter median hospital stay (VAN = 16 days (11–23), LIN = 12 (8–18) p = 0.09). In-hospital mortality was not significantly different between the groups (VAN 10.9%, LIN 8.7%, p = 1). Conclusion: Antibiotic escalation to Linezolid in febrile neutropenia following chemotherapy required significantly less additional antibiotic switches when compared to Vancomycin. Considering the good tolerability (especially the absence of significant renal side effects) and a comparable efficacy in this cohort, Vancomycin still represents a potent and more cost-effective option for suspected gram+ infection in patients with neutropenic fever. Disclosure: No conflict of interest disclosed. P247

A retrospective analysis of the impact of VRE colonization on patients with hematological malignancies Bodden G.1, Kondakci M.1, MacKenzie C.2, Kolbe-Busch S.2, Schulze-Röbbecke R.2, Germing U.1, Kobbe G.1, Fenk R.1, Schroeder T.1, Haas R.1 Uniklinik Düsseldorf, Klinik für Hämatologie, Onkologie und klinische Immunologie, Düsseldorf, Germany, 2Uniklinik Düsseldorf, Institut für Medizinische Mikrobiologie und Krankenhaushygiene, Düsseldorf, Germany 1

Introduction: The spread of vancomycin-resistant enterococci (VRE) strains causing infections may be associated with greater morbidity, mortality and healthcare cost. In this study, we evaluated the rates of VRE in patients with hematological malignancies on the basis of routine screening cultures obtained by rectal swabs. Methods: We routinely perform VRE screenings since 2009 with rectal swabs aiming to detect all VRE-colonized hematological patients treated in our Department of Hematology and Oncology. For this study, baseline data, rates of VRE infections and outcome data of colonized patients were collected. Analyzing the impact of colonization on patients’ clinical outcomes, we matched VRE-colonized patients with AML with AML patients not being found VRE-positive. Results: Between 2009–2013, a total of 1433 samples were examined, 330 (23,02%) of these were detected as VRE-positive according to 223 hematological patients, 133 (59,64%) male, 90 (40,36%) female, median age was 60 years (range 18–83 years), median length of hospital stay was 25 days (1–194) and median survival after detection of VRE was 291 (1–2093) days. The diagnoses were 86 cases of AML/MDS (38,57%), 71 NHL/ Hodgkin´s lymphomas (27,36%), 38 multiple myelomas (MM) (17,04%), 14 ALL (6,28%) and 24 other malignancies (10,78%). We compared the outcome of patients with VRE-findings and no VRE-findings in the subgroup of AML patients. One year survival rates were 77,5% (55 out of 71) and 74,6% (53 out of 71) respectively (p = 0,839). The rates of discontinuation of AML therapy were 16,41% and 21,09% respectively (p = 0.273). A total of 5 patients out of 223 (2,41%) with VRE developed VRE blood stream infections (BSI). Four of these were suffering from advanced hematological malignancies refractory to chemotherapy (1x AML, 1x AML + melanoma, 2 x NHL). These four patients died 1–15 days after VRE detection in blood cultures. One patient was in first line treatment of a MM. In this case VRE-BSI occurred in neutropenia and resolved without complications after treatment with Linezolid. Conclusions: In our single-center study the baseline characteristics of patients with VRE colonisation show a representative cross section of all patients with hematological malignancies. VRE colonisation did not show any significant influence on the outcome of patients with AML. VRE-BSI mainly occur in patients with end stage malignant diseases. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

P248

Computerized routine screening for malnutrition in cancer patients enables early nutritional interventions Stangl W.M.1, Stimpfl I.2, Guger-Halper U.3, Huber M.1, Szedlak G.1, Stangl T.A.4, Amtmann R.2 KH Oberwart, Hämato-/Onkologie und Palliativmedizin, Oberwart, Austria, KH Oberwart, Diätologie, Oberwart, Austria, 3KH Oberwart, Anstaltsapotheke, Zentrale Zytostatika-Aufbereitung, Oberwart, Austria, 4VU University, Amsterdam, Netherlands 1 2

Introduction: The prevalence of malnutrition in hospital patients in Europe lies between 20–50%, but in cancer patients this number goes up to 85%. About 20% of cancer patients die from the consequences of malnutrition and not from the tumour itself. According to a global survey conducted in 2012, 59% of cancer patients experienced weight loss >5% in the last 3 months, but only 8% received dietary counselling. Methods: Based on the recommendations of the European Society for Nutrition (ESPEN), the parameters of body weight, height and weight loss in the last 3 months were entered as a mandatory field in the record sheet of cancer patients. When a threshold is exceeded by 3 points, the underlying disease and the presence of additional stressors such as surgery, pressure ulcers or concurrent chemotherapy/radiotherapy or the presence of infection must be entered too. This means that with a total number of 3–5 points there is a risk of malnutrition, if the number is >5 a manifest malnutrition exists. In both cases, dietary counselling and specific therapy is automatically requested.The programming of the individual steps were made in i.s.h.med (SIEMENS AG). Results: During the observation period of 12 months, 237 patients were screened repeatedly for malnutrition (1488 patient cases). In 17 patients (7.17%) there was a score of 3–5 points (risk for malnutrition), and in 35 patients (14.77%), a score of 6 points or more (overt malnutrition). All patients with a score of 3 points or more( n = 52 (21,9%) received an initial consultation on the same day by the dietician and further nutritiontherapy planning. Conclusion: Our pilot study shows that all cancer patients treated in our institution are recognized as malnourished, or at risk for it, with the establishment of a computerized screening program. The risk or presence of malnutrition was detected according to the current ESPEN guidelines and targeted nutrition counselling and nutritional intervention were initiated promptly. Disclosure: No conflict of interest disclosed. P249

A retrospective analysis on the complications of intravenous port catheters in haemato-oncological patients Georgoula L.1, Schlicht E.2, Hebart H.3 Stauferklinikum Schwäbisch Gmünd, Klinik für Kinder- und Jugendmedizin, Mutlangen, Germany, 2Stauferklinikum Schwäbisch Gmünd, Klinik für Gynäkologie, Mutlangen, Germany, 3Stauferklinikum Schwäbisch Gmünd, Zentrum für Innere Medizin, Mutlangen, Germany 1

Introduction: Totally implantable port systems allow a simple venous access and are frequently used in haemato-oncological patients. The port chamber can be connected either to an open ended or a valved Groshong catheter. Methods: In a retrospective analysis we enrolled 294 consecutive patients who underwent a central venous port implantation between 2008–2011. The implanted catheter type and the frequency of catheter-related complications were analysed. In this analysis we compared the type of complications according to the type of implanted catheter. Results: Overall 310 port systems were implanted in 294 consecutive patients (162 female, 132 male) with an average age of 64,2 years (31–91 years). A system with an open-ended catheter was used in 207 patients (70,4%) whereas a Groshong catheter was used in 87 (29,6%) cases. Open-ended catheters were inserted into 193 patients suffering from solid tumours, 13 patients from haematological malignancies, and one patient

Abstracts

with a benign disease. Groshong catheters were inserted exclusively in female patients suffering from breast or gynaecological cancers. The total number of catheter-indwelling-days was 123.483 days. Systems with an open-ended catheter remained in situ for a total of 69.593 days (average 325 days), Groshong catheters for 53.890 days (average 561 days). Overall, complications related directly to the intravenous device occurred in 45 events (14,56%) in 36 patients. In the group of open-ended catheters we observed adverse events in 12,5% (n = 26) of the patients consisting of infectious complications (5,8%, n = 12), thrombotic complications in 1,9% (n = 4), postoperative wound bleeding in 1,4% (n = 3) and dislocation of the catheter in 1% (n = 2). Paravasation injury, pneumothorax and loosening of the port chamber occurred in 0,5% (n = 1) each. In 2 (1%) patients the implantation of the device was unsuccessful. In the Groshong-group we observed 19 complications (21,8%) consisting of thrombosis (6,9%, n = 6), dislocations of the catheter (5,7%, n = 5), infectious complications (3,45%, n = 3) and paravasation injury (3,45%, n = 3). Pneumothorax and wound bleeding were observed in 1,1% (n = 1) each. Conclusions: Totally implantable port-systems are safe and complication rates are low. In this retrospective study we could not observe any definite advantage of the Groshong-catheter when compared to open-ended catheters. Disclosure: No conflict of interest disclosed. P250

Development and validation of a novel real-time PCR assay for the rapid detection of fungal infections Rahn S.1, Schuck A.2, Kondakci M.2, Haas R.2, Pfeffer K.1, Henrich B.1 University of Düsseldorf, Department of Medical Microbiology and Hospital Hygiene, Düsseldorf, Germany, 2University of Düsseldorf, Department of Hematology, Oncology and Clinical Immunology, Düsseldorf, Germany 1

Introduction: Fungal infections are a major problem in immunocompromised patients, especially as they are difficult to diagnose due to low sensitivity and specificity of available tests. Detection of fungal pathogens by polymerase chain reaction (PCR) has recently become a promising diagnostic tool, although most of them specialize only on few selected fungal species. In this study, a real-time PCR assay was developed consisting of eight multiplex reactions for the standardized, broad range detection and differentiation of fungal pathogens in human specimens. Methods: The PCR targets the ITS1-ITS2 region of the fungal rDNA gene. Branch-specific sequence regions were identified in multiple sequence alignments and used to design primers for seven real-time PCRs covering distinct fungal branches, in addition to a pan fungal reaction. Positive reactions were detected by subsequent melting curve analysis and the fungal species were identified by Sanger sequencing of the respective PCR-product. Each Real time PCR reaction was validated by testing DNA of respective ATCC strains.121 throat swabs, 119 EDTA and 128 serum samples obtained from 51 patients with hematological malignancies during neutropenia were evaluated prospectively with the PCR assay and the results were viewed in the context of clinical findings. Results: Using this novel set of PCR assays, 368 samples of immune-compromised patients were analyzed. Tab. 1. PCR results

PCR results

throat swabs

EDTA

serum

samples

121

119

128

positive

67 (55,4%)

6 (50%)

10 (7,8%)

negative

54 /44,6%)

113 (95%)

118 (92,2%)

Overall, 16 different fungal species were detected with 49.4% (41/83) of the fungi belonging to Candida spp., 19 to Saccharomyces spp., 11 to Cladosporium spp. and 11 to other fungi of clinical relevance. Six of eight patients with highly suggestive signs of fungal pneumonia on high resolu-

Oncol Res Treat 2015;38(suppl 5):1–270

75


CONTENTS AUTHOR INDEX

tion CT had positive findings of Candida sp., Cladosporium sp. and two rarely reported fungi such as Pleosporales and Microdochium. Conclusion: In this study, a standardized broad range Real time PCR was validated to improve the diagnosis of invasive fungal infections in terms of speed and specificity. At this point, it is possible to identify the fungal genus of a patient sample in a single PCR run. Different methods of DNA extraction will be assessed in further studies to increase the sensitivity of the PCR in patients with hematological malignancies. Disclosure: No conflict of interest disclosed. P251

Clostridium difficile infections after autologous and allogeneic blood stem cell transplantation – a single center experience Kobbe G.1, Nagorny N.1, Mackenzie C.2, Rachlis E.1, Dienst A.1, Lopez Niedenhoff D.1, Schroeder T.1, Fenk R.1, Pfeffer K.2, Haas R.1, Kondakci M.1 Uniklinik Düsseldorf, Klinik für Hämatologie, Onkologie und klinische Immunologie, Düsseldorf, Germany, 2Uniklinik Düsseldorf, Institut für medizinische Mikrobiologie und Krankenaushygiene, Düsseldorf, Germany 1

Patients after blood stem cell transplantation (BSCT) are at risk of opportunistic and nosocomial infections. Among these, symptomatic Clostridium Difficile Infection (CDI) is of special importance because it is the most common nosocomial, antibiotic-associated infection. Between 1/2006 and 10/2014 a total number of 4797 stool specimens were analyzed for C.-diff. toxin from 751 patients with diarrhea who had previously received an auto or allo BSCT. Sixty-five individuals (MM 25, AML 23, Lymphoma 9, MDS/MPS 7 and 1 germ cell tumor) tested positive, reflecting an infection rate of 8.7% among all BSCT recipients with diarrhea (4,01% for autoTx and 8,16% for alloTx). Five patients (8%) had a history of CDI before Tx. The detection rate varied between 4.1% in 2012 and 10% in 2008 (mean 6.3%). When considering only the 1st year after BSCT, infection occurred earlier after autoTx (median day +37, range 1–331) than after alloTx (median day +85, range 3–364, p = 0.014) and in MM patients (median day +19, range 1–331) than in Lymphoma patients (median day +93, range 59–159, p = 0.03) which may be the result of metronidazole prophylaxis in allografted patients until day +20 after transplant and different previous antibiotic exposure. Of 29 autoTx patients with CDI 21(72%) relapsed with their primary malignancy (11 before, 9 after CDI). Of 36 alloTx patients 18(50%) relapsed (9 before, 9 after CDI). As therapy for CDI 40 patients (61.5%) received metronidazole, 12 (18.5%) vancomycin, 2 (3.1%) fidaxomycin, 7 (10.8%) unknown and 3 (4.6%) no therapy. No patient died of CDI but 16 (24.6%) had at least one recurrence (12 after metronidazole, 1 after vancomycin, 1 after fidoxomycin and 2 after unknown therapy). At day +30 after the 1st positive CDI test 93.1% and 94.5% of patients were alive after autoTx and alloTx, respectively. While overall mortality in CDI patients was similar after autoTX (55%, median FU 202 days, 4–2361) and alloTx (53%, median FU 365 days, 1–3223), causes of death differed (TRM 7%, DRM 48% after autoTX vs TRM 17%, DRM 36% after alloTx). Between 2006 and 2013 the rate of CDI among patients with diarrhea after BSCT varied from 4.1 to 10% and was related to diagnosis and type of transplant. CDI recurred frequently, especially after metronidazole. While CDI itself was not fatal, patients who developed CDI after blood stem cell transplantation resembled a high risk population with high treatment and disease related mortality. Disclosure: No conflict of interest disclosed.

P252

Management of chemotherapy-induced symptomatic anemia with epoetin theta as documented in the multicentre, non-interventional study EPO-amb Link H.1, Lück A.2, Rauh J.3, Bòrquez D.4, Maas C.5, Hesse J.6, Koenigsmann M.7, Schulze M.8, Tölg M.9, Scheuerlein R.W.10 Westpfalz-Klinikum Kaiserslautern, Klinik für Innere Medizin I, Kaiserslautern, Germany, 2Zentrum für Urologie und Onkologie, Rostock, Germany, 3 Fachinternistische Gemeinschaftspraxis und Therapiezentrum, Witten, Germany, 4Praxis für Onkologie und Hämatologie, Bergisch Gladbach, Germany, 5Gemeinschaftspraxis für Hämatologie und internistische Onkologie, Halberstadt, Germany, 6Gemeinschaftspraxis, Parchim, Germany, 7 Onkologisches Ambulanzzentrum OAZ, Hannover, Germany, 8Praxis für Urologie, Andrologie und Onkologie, Markkleeberg, Germany, 9Mediveritas GmbH, München, Germany, 10Teva GmbH, Berlin, Germany 1

Introduction: Chemotherapy-induced anemia (CIA) in cancer patients correlates with poor performance status and decreased quality of life. Currently by guidelines recommended therapies are erythropoiesis-stimulating agents, Fe substitution or a combination of both; RBC transfusions in case Hb-levels below 8–7g/dl. Material and methods: NIS EPO-amb is a multicenter, prospective observational study in Germany. 500 adult cancer patients (pts) with CIA treated by epoetin theta (Epo) were planned to be enrolled in 60 sites. We report on an interim analysis of the study. Results: 310 pts were enrolled by 07/2014. Mean (SD) age of pts was 63.6 (11.3) years with 64% females. The majority of pts had breast cancer (33%), followed by lung cancer (12%) and ovarian cancer (9%). Almost 2/3 of pts underwent chemotherapy with palliative intent. The following results are based on 162 pts with completed CIA treatment (max. over 12 weeks). CIA management: In addition to Epo 66 pts (40.7%) received transfusions, 28 pts (17.3%) iv Fe and 8 pts (4.9%) oral Fe substitution, resulting in the following CIA-management: Tab. 1.

CIA-management strategy

Patients

Percent (%)

Epo only

75

46.3

Epo + RBC transfusion

51

31.5

Epo + Fe iv

16

9.9

Epo + Fe oral

5

3.1

Epo + RBC transfusion + Fe iv

12

7.4

Epo + RBC transfusion + Fe oral

3

1.9

Hb-level: Median (Q1 / Q3) hemoglobin level at baseline (week 1) was 9.7 (9.0/10.3) g/dl . Overall, 93.8% of pts had Hb levels below 12.0 g/dl, 69.8% below 10.0 g/dl. Median Hb-level (Q1 / Q3) increased to 10.3 (9.5/11.2) in week 5 and 10.6 (9.8/11.7) in the last week of Epo treatment (or last Hb-value available). Epo dosage: Starting dose was 20.000 I.U. in 86.4% of all pts (10.000 I.U: 3.9%, 30.000 I.U: 9.3%, 40.000 I.U.: 0.4%). Only for 23 pts (14.2%) dosage was increased during the observational period. Mean (SD) duration of Epo treatment was 7.9 (3.6) weeks (median: 8.0 weeks). RBC transfusions: For 66 pts (40.7%) a total of 99 RBC transfusions was reported. Overall, in 78.3% of transfusions the Hb-value at baseline was >8.0 g/dl. Conclusions: EPO-amb data show that epoetin theta is mainly used according to the labeled recommendation to start with 20.000 I.U. A high proportion of patients received transfusions while this was not justified. Use of guidelines for treatment of CIA should be improved. Disclosure: Hartmut Link: Advisory Role: Teva, Amgen, Hexal, Vifor-Pharma; Financing of Scientific Research: Teva, Amgen, Hexal, Vifor-Pharma; Expert Testimony: Amgen. Robert Willy Scheuerlein: Employment or Leadership Position: TEVA GmbH.

76

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

P253

Privigen® in secondary immunodeficiencies – interim analysis of a multicenter non-interventional study in Germany Plath M.1, Slawik H.R.1, Reiser M.2, Otremba B.3, Pfründer D.4 Onkologische Schwerpunktpraxis, Augsburg, Germany, 2Praxis internistischer Onkologie und Hämatologie, Köln, Germany, 3Onkologische Praxis, Oldenburg, Germany, 4CSL Behring, Hattersheim, Germany 1

Introduction: Privigen® is a 10% liquid preparation of polyvalent human IgG for intravenous administration. The use of the stabiliser L-proline fully preserves IgG functional activity without refrigeration, making Privigen® ready-to-use. Privigen® is licenced as a maintenance therapy in primary and secondary immunodeficiencies, and as an immunomodulatory therapy in autoimmune and inflammatory diseases. Methods: This is an interim analysis of an ongoing multicenter non-interventional study to evaluate the efficacy and tolerability of Privigen®, focussing on secondary immunodeficiencies. The cut-off date was March 8, 2015. Results: 1,554 patients (824 males, 730 females; mean age 67 years) in 145 centers received a total of 18,128 Privigen® infusions. The mean observation period was 12.5 months. The average monthly dose was 14 g (0.2 g/ kg body weight). Underlying diseases with n≥5 were chronic lymphocytic leukemia (n = 766), acute lymphocytic leukemia (n = 8), chronic myeloid leukemia (n = 5), acute myeloid leukemia (n = 22), hairy-cell leukemia (n = 5), Hodgkin lymphoma (n = 19), non-Hodgkin lymphoma (n = 373), myeloma (n = 282), macroglobulinemia Waldenström (n = 42), monoclonal gammopathy (n = 21), myelodysplastic syndrome (n = 11) and solid tumor (n = 22). The efficacy was judged as very good or good in 94.4%, moderate in 4.7% and insufficient in 0.9% of evaluable cases (n = 1,439). Patients who had not received any IVIG treatment prior to study entry and who received ≥6 infusions of Privigen® for ≥120 days (infusion intervals ≥20 d and ≤60 d) experienced significantly fewer infections during the study than before: The mean annualized infection rate dropped from 5.4 to 1.6 (n = 143; p < 0.0001). The tolerability was judged as very good or good in 92.7%, moderate in 4.8% and insufficient in 1.9% of all cases (0.6% missing data). Adverse events possibly or probably related to Privigen® were reported for 243 of the 18,128 infusions (1.3%); 17 events were considered serious (0.1%). Conclusions: Privigen® significantly reduced infection rates in secondary immunodeficiencies. The tolerability was very good or good in the majority of patients. Disclosure: Margarete Plath: Financing of Scientific Research: Ja. Dietmar Pfründer: Employment or Leadership Position: Ja.

id and liposomal amphotericine B. The patient deteriorated rapidly with respiratory failure due to ARDS. BAL revealed a significant viral load of HCoV-NL63 (Ct = 30 in real-time RT-PCR, no other respiratory viruses detected). Based on successful application of pegylated IFN against the related SARS-coronavirus in animal experiments, a single injection of 180µg PEG-IFN-α2b was applied. Intravenous immunoglobulins were given because of the wide prevalence of anti-HCoV-NL63 in the population. Steroids were provided to limit inflammation, based on experience with SARS-CoV. Despite immediate initiation of treatment and elimination of virus in subsequent tests, septic shock with progressive lung failure led to death 7 days after onset of fever with massive lung bleeding as a consequence of diffuse alveolar haemorrhage. Conclusion: This report emphasizes the fatal consequences of common respiratory virus infections in immunocompromized patients. HCoV which in total account for 5–10% of adult upper respiratory tract infections can be blocked by treatment with peg-IFN if diagnosed early. Disclosure: No conflict of interest disclosed. P255

Prophylactic treatment of chemotherapy-induced neutropenia with Lipegfilgrastim in patients with breast cancer: subgroup analysis of the non-interventional study NADIR Kurbacher C.M.1, Fietz T.2, Salat C.3, Zaiss M.4, Gazawi N.5, Steffens C.-C.6, Egert M.7, Graffunder G.8, Papke J.9, Weißenborn G.10, Bückner U.11, Illmer T.12, Jungberg P.13, Lorenz A.14, Weide R.15, Klare P.16, Tesch H.17, Oskay-Özcelik G.16, Teichmann B.18, Harde J.18, Scheuerlein R.19 Gynäkologisches Zentrum Bonn-Friedensplatz – Schwerpunkt gynäkologische Onkologie, Bonn, Germany, 2Schwerpunktpraxis für Innere Medizin, Hämatologie, Onkologie und Gastroenterologie, Singen, Germany, 3 Hämato-Onkologische Schwerpunktpraxis, München, Germany, 4Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany, 5Klinik für gynäkologische Onkologie, Leipzig, Germany, 6MVZ für Hämatologie/Onkologie Klinik Dr. Hancken, Stade, Germany, 7Praxis für Innere Medizin, Hämatologie, Intern. Onkologie, Werdau, Germany, 8Frauenarzt-Zentrum-Zehlendorf, Berlin, Germany, 9Internistische Praxis und Tagesklinik, Neustadt i.S., Germany, 10Praxis für Hämatologie und Onkologie, Twistringen, Germany, 11Hämatologischonkologische Schwerpunktpraxis, Bochum, Germany, 12Gemeinschaftspraxis Hämatologie – Onkologie, Dresden, Germany, 13Gynäkologische Praxis, Chemnitz, Germany, 14Frauenarztpraxis, Hildburghausen, Germany, 15Praxis für Hämatologie und Onkologie, Koblenz, Germany, 16Praxisklinik Krebsheilkunde für Frauen / Brustzentrum, Berlin, Germany, 17Hämatologisch-Onkologische Gemeinschaftspraxis am Bethanien-Krankenhaus, Frankfurt a. M., Germany, 18 iOMEDICO AG, Freiburg, Germany, 19TEVA GmbH, Berlin, Germany 1

Introduction: Human Coronavirus NL63 (HCoV-NL63) is one of four common respiratory CoV. Despite high incidence and options for treatment with interferon, HCoV infections are grossly understudied. We here report a case of HCoV infection in a leukemia patient with fatal ARDS despite successful virus elimination by pegylated Interferon-alpha (PEGIFN-α). Case: The 27-year old female pre-T-ALL patient was treated according to the German-Multicenter Trial for Adult ALL (GMALL) 07–03 protocol. No relevant infectious complications were seen until day 35 when the neutropenic patient developed fever without any clinical infectious focus. Antibiotic prophylaxis was switched to meropenem. Two hours later the patient collapsed, a positive shock index immediately triggered ICU referral. Anti-infectious therapy was escalated to additional linezol-

Introduction: Anthracycline and/or taxane-based chemotherapy regimen are the most effective therapies in breast cancer. Chemotherapy-induced neutropenia (CIN) and febrile neutropenia (FN) are two of the most common and life-threatening toxicities of myelosuppressive chemotherapy regimen leading to dose reductions and delay of cycle intervals. Corresponding guidelines recommend primary prophylaxis with granulocyte colony-stimulating factors (G-CSF) in patients receiving chemotherapy with a high risk for febrile neutropenia (FN). Lipegfilgrastim is a glyco-pegylated r-metHuG-CSF approved to reduce the duration of neutropenia and the incidence of FN. We report on NIS NADIR looking at systemic treatment and use of Lipegfilgrastim in breast cancer pts. Methods: This prospective NIS was initiated to collect data on prophylactic use of lipegfilgrastim in patients at risk for CIN according to the corresponding guidelines. As of 12/2013 a total 120 outpatient centers and hospitals in Germany are planned to recruit 1.500 patients (pts). Data on demographics, tumour characteristics, comorbidities, previous and ongoing systemic treatments, and Lipegfilgrastim treatment are collected. Pts complete a customized questionnaire focusing on self-injection and handling of the syringe. Main objectives are the incidence of severe neutropenia (NCI CTCAE grade 3–4), FN and resulting complications. Results: At time of the interim analysis (04/2015) 80 sites enrolled 835 pts. 717 pts were evaluable thereof 317 pts with breast cancer. Mean age was 54.8 (SD: ±11.2) years. At inclusion 89.9% had an ECOG status of 0 or 1.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

P254

Fatal outcome of human coronavirus infection despite successful viral elimination by IFN-alpha in a patient with newly diagnosed ALL Mayer K.1, Nellessen C.1, Wallau A.1, Schumacher M.1, Pietzonka S.2, Drosten C.2, Brossart P.1, Wolf D.1 Universitätsklinik Bonn, Medizinische Klinik III, Bonn, Germany, Universitätsklinik Bonn, Institut für Virologie, Bonn, Germany

1 2

77


CONTENTS AUTHOR INDEX

Chemotherapy was mostly given in (neo) adjuvant setting (80.8%). Overall 94.6% pts were treated with taxane and/or anthracycline combinations. 23.3% were treated with dose-dense regimen (q2w regimen including epirubicine and/or cyclophosphamide and/or taxanes). Lipegfilgrastim was mostly given within 3 days after chemotherapy application (93.9%). Nearly all pts treated with dose-dense chemotherapy received Lipegfilgrastim within 3 days after chemotherapy (96%). 1.3% (n = 4) developed FN during the first Lipegfilgrastim supported cycle, thereof one pt (0.3%) received a dose-dense regimen. Conclusions: These data show, that in a primary (neo) adjuvant setting with mostly anthracycline and/or taxane-based chemotherapy, application and efficacy of Lipegfilgrastim was as expected. The low incidence of FN was comparable to literature. Disclosure: Christian Martin Kurbacher: Advisory Role: TEVA GmbH; Financing of Scientific Research: TEVA GmbH. Robert Scheuerlein: Employment or Leadership Position: TEVA GmbH.

during the first Lipegfilgrastim supported cycle. Twelve pts (13.2% [95%CI: 7.0–21.9%]) developed NCI CTCAE grade 3 or 4 neutropenia in the first Lipegfilgrastim supported cycle. Conclusions: Here for the first time data to the application of Lipegfilgrastim in hematological diseases are shown. In a mostly R-CHOP-based chemotherapy setting, application and efficacy of Lipegfilgrastim was as expected. The low incidence of FN is comparable to literature. Disclosure: Thomas Fietz: Advisory Role: TEVA GmbH; Financing of Scientific Research: TEVA GmbH; Other Financial Relationships: TEVA GmbH Robert Willy Scheuerlein: Employment or Leadership Position: TEVA GmbH. P257

Fosfomycin in febrile neutropenic patients with haematological malignancies: A retrospective case documentation Schwab K.S.1, Mayer K.1, Brossart P.1, Hahn-Ast C.1 Universitätsklinik Bonn, Medizinische Klinik III, Bonn, Germany

1

P256

Prophylactic treatment of chemotherapy-induced neutropenia with Lipegfilgrastim in patients with NHL: Subgroup analysis of the non-interventional study NADIR Fietz T.1, Wolff T.2, Sandner R.3, Janssen J.4, Hurtz H.-J.5, Heits F.6, Weide R.7, Weißenborn G.8, Losem C.9, Lück A.10, Kurbacher C.M.11, Teichmann B.12, Harde J.12, Scheuerlein R.W.13 Schwerpunktpraxis für Innere Medizin, Hämatologie, Onkologie und Gastroenterologie, Singen, Germany, 2OncoResearch Lerchenfeld GmbH, Hamburg, Germany, 3Gemeinschaftspraxis für Hämatologie und Internistische Onkologie, Passau, Germany, 4Gemeinschaftspraxis für Hämatologie und Onkologie, Westerstede, Germany, 5Onkologische Gemeinschaftspraxis, Halle (Saale), Germany, 6Agaplesion Diakonieklinikum Rotenburg gGmbH, Rotenburg (Wümme), Germany, 7Praxis für Hämatologie und Onkologie, Koblenz, Germany, 8Praxis für Hämatologie und Onkologie, Twistringen, Germany, 9Praxis für Onkologie und Hämatologie, Neuss, Germany, 10Zentrum für Urologie und Onkologie, Rostock, Germany, 11Medizinisches Zentrum Bonn-Friedensplatz – Schwerpunkt gynäkologische Onkologie, Bonn, Germany, 12iOMEDICO AG, Freiburg, Germany, 13TEVA GmbH, Berlin, Germany 1

Introduction: Chemotherapy-induced neutropenia (CIN) and febrile neutropenia (FN) are serious dose-limiting toxicities. Lymphoma patients who develop FN are typically hospitalized and treated with intravenous antibiotics. As a consequence the dose of chemotherapy is often reduced and/or the cycle intervals are prolonged, always associated with poor outcomes. This is why existing guidelines recommend the use of G-CSF in primary prophylaxis for patients (pts) at high FN risk or for pts at intermediate risk in the presence of additional risk factors. Lipegfilgrastim is a glyco-pegylated r-metHuG-CSF approved to reduce the duration of neutropenia and the incidence of FN. We report on interim data obtained during the course of the non-interventional study (NIS) NADIR in NHL patients. Methods: This prospective NIS NADIR was initiated to collect data on prophylactic use of Lipegfilgrastim in patients with high risk for CIN according to the corresponding guidelines. A total of 120 outpatient centers and hospitals in Germany are planned to recruit 1.500 pts. Data on demographics, tumour characteristics, comorbidities, previous and ongoing systemic treatments, and Lipegfilgrastim treatment are collected. Main endpoint is the incidence of severe neutropenia (NCI CTCAE grade 3–4), FN and resulting complications. Results: At time of the interim analysis (04/2015) 80 sites in Germany enrolled 835 pts. 717 pts were evaluable with at least one completely documented Lipegfilgrastim cycle. The 91 pts with NHL are discussed here: Mean age was 64.9 (SD ± 12) years, with 67% males. At inclusion 81.4% of all pts had an ECOG status of 0 or 1 (with 16.5% missing data). Overall 39.6% pts were treated with (R)-CHOP, (R)-CHOP-like (24.2%) and (R)-Bendamustin (13.2%). Lipegfilgrastim was mostly given within 3 days after chemotherapy application (88%). For 7.7% (n = 7; CI 3.1–15.2%) dose modification was reported after the first Lipegfilgrastim supported cycle, thereof 2 cases (2.2%) due to neutropenia. No pts developed FN

78

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: Fosfomycin (FOS) is a phosphonic acid derivative with bactericidal activity and an expanded spectrum of activity. There is no published experience of treating invasive infections in onco-hematological patients. We observed that occasionally critically ill neutropenic patients unresponsive to other antibiotics were treated with FOS in our departments. Aim of our study was to analyse effectiveness and toxicity of FOS in febrile neutropenia (FN). Methods: Data of febrile neutropenic episodes treated with FOS were retrospectively collected. Baseline data of the patients, therapy of haematological malignancy, treatment of infection and adverse events were documented. Success was defervescence (>/= 7d) in absence of any sign of continuing infection. Results: Data of eighteen patients (12 female, 6 male) were collected so far. Median age was 57 years (range 26–75 years). All patients (pts.) had haematological malignancies (12 AML, 2 ALL, 2 NHL, 2 Multiple Myeloma). All pts. were neutropenic at the onset of fever with a median duration of 19 days (d) (range 5–40 d). The type of infection was pneumonia in 3 pts., 14 Bacteriaemia, 1 FUO. Before initiation of FOS, 8 pts. (44%) received two lines or more of antibiotic therapy. FOS (15g/d) was given for a median of 10 d (range 3–25 d). In 6/16 pts. an invasive fungal infection was considered as possible and in two as probable. A viral infection was never assumed. Treatment was successful in 10/16 pts. (56%), in two patients success of FOS could not be evaluated. Four pts. (22%) died due to infection. Grade 3–4 toxicity did not occur. Conclusions: Although this analysis included very ill patients, our results showed promising response rates to FOS. Since success rates of escalated antibiotic therapy in FN are generally very low, we consider FOS a promising alternative for salvage antibiotic therapy in FN. This case documentation will be continued to get a more precise idea of the efficacy of FOS in FN. Disclosure: No conflict of interest disclosed. P258

Multiprofessional cancer medication management at the center for integrated oncology Jansen C.1, Zipfel M.2, Koch L.2, Becker G.2, Schulze I.3, Kuhn W.4, Schmidt-Wolf I.G.H.2, Jaehde U.1 Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany, Center for Integrated Oncology Cologne/Bonn, Department of Internal Medicine III, University Hospital, Bonn, Germany, 3Hospital Pharmacy, University Hospital, Bonn, Germany, 4Department of Obstetrics and Gynecology, Center for Integrated Oncology Cologne/Bonn, University Hospital, Bonn, Germany 1 2

Introduction: Due to the high toxicity of anti-cancer drugs and the complexity of the medication, patients are at high risk of experiencing adverse events. Patient safety may be enhanced by providing a structured multiprofessional medication management for cancer patients.

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

Methods: The medication management consists of four multiprofessional care modules composed of a basic module (medication review, interaction check) and specific modules for the management of three common adverse events (nausea and emesis, mucositis, fatigue). All care modules contain a care algorithm, evidence-based recommendations for supportive care and patient information brochures. The medication management was tested in a single-arm pilot study by measuring patient-reported symptom load, quality of life and patient satisfaction with information. Results: For the pilot study 21 cancer patients with any solid tumor were recruited. The results show the feasibility of the medication management and acceptance by the patients and the multiprofessional care team. The most frequently applied adverse event module was nausea and emesis (100%) followed by mucositis (91%) and fatigue (62%). Mainly patients with head and neck cancer receiving concomitant radiochemotherapy showed early toxic symptoms of high severity. Conclusion: Our pilot study confirmed the feasibility and acceptance of structured multiprofessional care. Assuming that patients with head and neck cancer may particularly benefit from medication management, a randomized two-arm study will be conducted by focusing on this patient cohort. Primary endpoint of the study will be the frequency and severity of treatment-associated toxicity. Disclosure: No conflict of interest disclosed.

Posterdiskussion

Urogenitale Malignome P259

FNTB promoter polymorphisms influence survival in prostate cancer Virchow I.1,2, Schmid K.W.3, Rübben H.4, Heukamp L.C.5,6, Siffert W.1, Bachmann H.S.1 Institute of Pharmacogenetics, Medical Faculty, University of Duisburg-Essen, Essen, Germany, 2West German Cancer Center, University Hospital Essen, Department of Medical Oncology, Essen, Germany, 3Institute of Pathology and Neuropathology,Medical Faculty, University of Duisburg-Essen, Essen, Germany, 4 Department of Urology, Medical Faculty, University of Duisburg-Essen, Essen, Germany, 5NEO New Oncology, Cologne, Germany, 6Hämatopathologie, Hamburg, Germany 1

Introduction: Farnesyltransferase inhibitors (FTIs) revealed promising results in vitro as antitumoral therapeutic strategy. They inhibit activation of different tumor related proteins. The reason why FTIs (Lonafarnib, Tipifarnib) failed in large clinical trials remains unclear. Predictive biomarkers to identify patients, who most likely benefit from FTIs would be highly useful. The primary target of FTIs is the farnesyltransferase β-subunit (FNTB). Therefore we functionally analysed polymorphisms in the promoter region of the FNTB gene. In addition the prognostic impact of these polymorphisms were evaluated in patients with prostate cancer. Methods: Gene sequencing including promoter regions were performed using the method by Sanger. Functional analysis of detected polymorphisms were evaluated by electrophoretic mobility shift (EMSA) and reporter assays. Two independent prostate cancer patient populations (n = 222, n = 179) were retrospectively genotyped for the polymorphisms using restriction enzymes and pyrosequencing. Association of genotypes as well as haplotypes with risk for cancer and disease progression were analysed using Cox regression and Kaplan- Meyer analysis. Results: Three promoter polymorphisms were detected, -609G/C, -179T/ A, and -173delG. EMSA analysis for the polymorphisms -609 and -173 revealed different binding properties of transcription factors, genotypeand haplotype-dependent differences of promoter activity and altered FNTB expression dependent on genotype. Kaplan Meyer analysis showed significant association of polymorphisms and haplotypes with survival in the combined analysis of the -609 and -173 polymorphisms (p = 0.021) in one population. Patients with the -173 del/del genotype combined with the -609 CC genotype show the worst survival, but the best survival if they

80

Oncol Res Treat 2015;38(suppl 5):1–270

carry at least one -609 G allele. In the second population the -609 CC genotype independently influenced disease free survival till metastasis. These patients had the worst overall survival (p = 0.006). Conclusions: In patients with prostate cancer functional FNTB promoter polymorphisms influence FNTB expression and survival. Our results suggest the FNTB polymorphisms as promising target for further evaluation with regard to their prognostic as well as predictive impact. Disclosure: No conflict of interest disclosed. P260

Quality of life (QoL) and patterns of care in second line treatment of metastatic castration resistant prostate cancer (mCRPC) after prior chemotherapy docetaxel chemotherapy: Results of a prospective Swiss observational treatment registry (SEQOND) Stenner F.1, Betticher D.2, Rothschild S.1, Caspar C.3, Morant R.4, Popescu R.A.5, Rauch D.6, Huber U.S.7, Zenhäusern R.8, Rentsch C.9, Cathomas R.10 Universitätsspital Basel, Onkologie, Basel, Switzerland, 2HFR Fribourg – Hôpital cantonal, Oncologie, Fribourg, Switzerland, 3Kantonsspital Baden, Medizinische Klinik/ Onkologie, Baden, Switzerland, 4ZeTuP Rapperswil-Jona, Tumorzentrum, Rapperswil, Switzerland, 5Hirslanden Medical Center, Tumor Zentrum, Aarau, Switzerland, 6Spital STS AG, Medizinische Onkologie, Thun, Switzerland, 7 Onko-Zentrum, Zürich, Switzerland, 8Spitalzentrum Oberwallis, Onkologie, Brig, Switzerland, 9Universitätsspital Basel, Urologie, Basel, Switzerland, 10 Kantonsspital Graubünden, Abteilung Onkologie und Hämatologie, Chur, Switzerland 1

Introduction: Several new treatment options have demonstrated improved overall survival in the treatment of mCRPC after prior chemotherapy with docetaxel including the cytotoxic agent cabazitaxel. In the pivotal phase III trial of cabazitaxel (TROPIC) no evaluation of QoL on treatment with cabazitaxel was performed. With a variety of second line options the optimal treatment sequence after prior docetaxel remains elusive. We prospectively evaluated quality of life of different treatments at progression after prior docetaxel and describe the patterns of care of second line treatment in Switzerland. Methods: Patients (pts) with mCRPC and disease progression after having received >225 mg/m2 docetaxel as first line treatment were eligible. Pts were enrolled consecutively in participating centers. Second line treatment consisted of cabazitaxel (25 mg/m2 every 3 weeks) or any other treatment including abiraterone acetate. Choice of second line treatment was at the discretion of the investigator. Primary objective was assessment of QoL maintenance under second line treatment. Secondary objectives included improvement of QoL, evolution of QoL as well as assessment of fatigue and pain during second line therapy. QoL was measured using the FACT-P questionnaire. QoL maintenance was defined as having at maximum a decrease in two functional domains of the FACT-P at any time point compared to baseline. Fatigue was evaluated with the BFI and pain with the MPQ-SF pain questionnaire. Questionnaires were completed at baseline and then monthly for four months. All pts gave their informed consent and an ethical approval was received for all participating centers. Results: A total of 165 pts were included in 35 Swiss centers between November 2011 and December 2014. 72 pts were treated with cabazitaxel and 93 pts with other second line treatments. The data presented on the primary and secondary endpoints at the time of the meeting will give a detailed overview of the treatment arms and the outcomes. Currently, the data are not mature since not all pts have completed their evaluation period at this time point. Conclusions: We will be able to show prospectively collected QoL data including fatigue and pain assessments on treatment with cabazitaxel and other second line treatment options. Moreover our data should give insight into the different usage of second line treatment options for pts with mCRPC and describe the patterns of care in Switzerland. The study was funded by Sanofi/CH.

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

Disclosure: Frank Stenner: Advisory Role: Nicht regelmässig, aber siehe 5.; Financing of Scientific Research: Im Rahmen von ad hoc advisory boards von Sanofi Honare für Beratertätigkeiten erhalten; Expert Testimony: Finanzielle Unterstützung für wissenschaftliche Projekte durch Sanofi; Other Financial Relationships: Reisekostenerstattungen und Kongressgebühren für Kongresse. Richard Cathomas: Advisory Role: Nicht regelmässig, aber siehe 5.; Financing of Scientific Research: Im Rahmen von ad hoc advisory boards von Sanofi Honare für Beratertätigkeiten erhalten; Expert Testimony: Finanzielle Unterstützung für wissenschaftliche Projekte durch Sanofi; Other Financial Relationships: Reisekostenerstattungen und Kongressgebühren für Kongresse. P261

Impact of prior docetaxel, Extent of Disease (EOD), and prior Bisphosphonates (Bp) on Hematologic (Heme) safety of radium-223 Dichloride (Ra-223) from ALSYMPCA Strauss A. , Michalski J.M. , Parker C. , Sartor O. , Vogelzang N.J. , Haugen I.6, Garcia-Vargas J.7, Nilsson S.8 1

2

3

4

5

Universitätsmedizin Göttingen, Urologische Klinik und Poliklinik, Göttingen, Germany, 2Washington University School of Medicine, St Louis, United States, 3 The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Sutton, United Kingdom, 4Tulane Cancer Center, New Orleans, United States, 5 Comprehensive Cancer Centers of Nevada, Las Vegas, United States, 6Algeta ASA, Oslo, Norway, 7Bayer HealthCare, Whippany, United States, 8Karolinska University Hospital, Stockholm, Sweden 1

Objective: Ra-223, a first-in-class α-emitter, reduced risk of death by 30% in phase 3 ALSYMPCA (Parker et al. NEJM 2013; ALSYMPCA) and is approved in 34 countries to date for treatment (tx) of CRPC with symptomatic bone mets and no known visceral disease. In ALSYMPCA, Gr 3 and 4 heme adverse events (AEs) incidence was low, with no clinically meaningful differences vs placebo (Pbo). This post hoc subgroup analysis evaluated whether prior docetaxel, prior Bp, or EOD predisposes pts with Ra-223 tx to myelosuppression. Methods: ALSYMPCA pts had progressive CRPC with ≥ 2 symptomatic bone mets and no known visceral mets, were receiving best standard of care, and previously received docetaxel or were unfit for or declined docetaxel. Pts (2:1) had 6 injections of Ra-223 (50 kBq/kg IV; q 4 wk) or matching Pbo. Subgroup analyses of Gr 3/4 anemia, neutropenia, and thrombocytopenia by prior Bp and EOD at baseline used Cox regression; a prespecified analysis determined heme AE incidences in docetaxel subgroups. Results: No tx differences were observed for Gr 3/4 anemia across all subgroups. Gr 3/4 thrombocytopenia was higher in Ra-223 vs Pbo pts for prior docetaxel, EOD > 20 mets/superscan, and prior Bp. For Ra-223, Gr 3/4 neutropenia was higher with prior vs no prior docetaxel; Gr 3/4 thrombocytopenia was higher with prior vs no prior Bp, EOD >20 mets/superscan vs ≤20 mets, and prior vs no prior docetaxel. For Pbo, Gr 3/4 anemia was higher for >20 mets/superscan vs ≤20 mets; Gr 3/4 thrombocytopenia was higher with prior vs no prior docetaxel. Conclusion: As in ALSYMPCA overall analyses, Gr 3/4 anemia showed no differences for Ra-223 vs Pbo across all subgroups of CRPC pts. Gr 3/4 neutropenia was higher for Ra-223 with prior vs no prior docetaxel. Thrombocytopenia was higher with Ra-223, prior docetaxel, greater EOD, and prior Bp. Disclosure: Arne Strauss: Financing of Scientific Research: Bayer, Pfizer, Novartis, Amgen. Sten Nilsson: Advisory Role: Bayer.

P262

Quality-of-life (QoL) of patients with metastatic castration resistant prostate cancer (mCRPC) treated with Cabazitaxel – results from the non-interventional study QoLiTime Al-Batran S.-E.1, Lange C.2, Ecke T.3, Windemuth-Kieselbach C.4, Kloss S.5, Hammerer P.6, Hofheinz R.-D.7 Hospital Nordwest, Institute of Clinical Research, Frankfurt, Germany, Practice for Urology and Uro-Oncology, Bernburg, Germany, 3Helios Hospital, Department of Urology, Bad Saarow, Germany, 4Alcedis GmbH, Biometry, Gießen, Germany, 5DRK Hospital, Center for Prostate Cancer, Luckenwalde, Germany, 6Academic Hospital Braunschweig, Department of Urology and Uro-oncology, Braunschweig, Germany, 7University Hospital Mannheim, Interdisciplinary Tumor Center, Mannheim, Germany 1 2

Background: Cabazitaxel (Caba) combined with Prednis(ol)one is approved for 2nd-line treatment of mCRPC after Docetaxel. Potential toxicity of chemotherapy may impact on patient´s QoL and counterbalance treatment benefits. Thus QoL data are important from physicians´, patients’ and regulatory perspective. Methods: Patients with mCRPC receiving Caba were included in the non-interventional study QoLiTime. EORTC QLQ C30 was handed out at baseline and each cycle. We present QoL results and the correlation of QoL with biochemical response (PSA decrease >50%) of 238 patients from baseline (BL) and after 4 cycles (C4) of Caba. Results: The total population consists of 527 patients, 406 received 4 cycles of Caba, 266 of those had PSA measurements at BL and C4 and 238 completed the EORTC QLQ 30 at BL and C4. Median age was 72 years with good performance status (ECOG 0:36%; ECOG 1:54%). No significant change in QoL was observed between BL and C4, independent of PSA response. At C4, there was an improvement in physical and role functioning (–5.21; p < 0.0001 and –8.79; p < 0.0001) while there was a worsening of fatigue (6.49; p = 0.0003), dyspnoea (6.36; p = 0.004) and diarrhoea (9.16; p < 0.0001). Other scales did not vary significantly from BL to C4. Improvement in physical functioning and pain decrease (–7.61) were associated with a PSA decrease >50%. Conclusions: This study is the largest prospective analysis of QoL in patients receiving Cabazitaxel for mCRPC. Symptom increases were seen in typical areas of chemo toxicity such as fatigue and diarrhoea but QoL was maintained during the 12-week observation period. Improvement in physical functioning and pain was associated with a good PSA response. This study is funded by Sanofi. Disclosure: Salah-Eddin Al-Batran: Financing of Scientific Research: Sanofi. Ralf-Dieter Hofheinz: Financing of Scientific Research: Sanofi; Expert Testimony: Sanofi. P263

Trials in progress: The influence of the therapy sequence in patients with metastatic castration resistant prostate cancer (mCRPC) treated with Cabazitaxel – the international, prospective non-interventional trial SCOPE Bokemeyer C.1, Stoiber F.2, Amram M.-L.3, Gschwend J.E.4 University Hospital Eppendorf (UKE), Department of Oncology, Hematology with section Pneumology, Hamburg, Germany, 2LKH Vöcklabruck, Department of Urology and Andrology, Vöcklabruck, Austria, 3Geneva University Hospital, Department of Oncology, Geneva, Switzerland, 4Klinikum rechts der Isar, Technische Universität München, Department of Urology, Munich, Germany 1

Introduction: In the treatment of patients with metastatic castration resistant prostate cancer (mCRPC) different options at progression after Docetaxel are available. However, clear evidence to decide which drug to use for which patient at what time point is not yet available. Retrospective data have led to the hypothesis that post-docetaxel, the sequence of Cabazitaxel followed by an androgen receptor (AR) targeted agent may be associated with a longer survival than the inverse sequence. The prospective, international, multicenter, non-interventional study SCOPE will evaluate the influence of the sequence of different drugs on the therapeutic out-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

81


CONTENTS AUTHOR INDEX

AUTHOR INDEX

come by comparison of results based on different Cabazitaxel treatment time points as used in daily clinical practice. Methods: The SCOPE study will be conducted in 300 centers in Germany, Austria and Switzerland with a total of 900 planned patients, starting in summer 2015. Patients who are started on Cabazitaxel treatment and who have received prior Docetaxel therapy will be consecutively enrolled. The primary objective is to evaluate the impact of treatment sequence on progression free survival (PFS) with cabazitaxel, as assessed by the investigator. The analysis will focus mainly on the following two sequences: 1. Docetaxel pre-treatment followed directly by Cabazitaxel (continuous taxane-therapy sequence), 2. Docetaxel followed by an AR-targeted or other therapy prior to Cabazitaxel (intermittent taxane-therapy sequence). Secondary endpoints include PSA response, time to PSA progression with each therapy, number of Cabazitaxel cycles received, overall survival and safety. Data of each patient will be collected prospectively from time of first dose of Cabazitaxel until 24 months follow-up after first dose of Cabazitaxel or until death. Conclusion: An important question for the optimal treatment of patients with mCRPC is the best treatment sequence according to patient and tumor characteristics. Selection of therapies may be influenced by potential resistance between AR-targeted therapies and patient performance status. SCOPE aims to analyze and compare outcomes for different sequences in which Cabazitaxel is used in daily clinical practice to gather insight into this important question. This study is funded by Sanofi. Disclosure: Carsten Bokemeyer: Advisory Role: Sanofi; Financing of Scientific Research: Sanofi. Jürgen E. Gschwend: Advisory Role: Sanofi; Financing of Scientific Research: Sanofi. P264

External-beam radiation therapy (EBRT) use and safety with Radium-223 dichloride (Ra) in patients (pts) with castrationresistant prostate cancer (CRPC) and symptomatic bone metastases (mets) from the ALSYMPCA trial Strölin P.1, O’Sullivan J.2, Sartor O.3, Parker C.4, Hoskin P.5, Widmark A.6, Mellado B.7, Helle S.8, Aksnes A.9, Garcia-Vargas J.10, Nilsson S.11 Martini-Klinik am UKE GmbH, Hamburg, Germany, 2Queen’s University, Centre for Cancer Research and Cell Biology, Belfast, United Kingdom, 3Department of Medicine: Section of Hematology & Medical Oncology and Department of Urology, Tulane Cancer Center, New Orleans, United States, 4Academic Urology, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Sutton, United Kingdom, 5Mount Vernon Hospital Cancer Centre, Middlesex, United Kingdom, 6Radiation Sciences, Oncology, Umeå University, Umeå, Sweden, 7Medical Oncology, Hospital Clinic, Barcelona, Spain, 8Oncology, Haukeland University Hospital, Bergen, Norway, 9Clinical Research, Algeta ASA, Oslo, Norway, 10Oncology Global Clinical Programs, Bayer HealthCare, Whippany, United States, 11Clinical Oncology, Karolinska University Hosiptal, Stockholm, Sweden 1

Aim: Bone mets in CRPC frequently cause symptomatic skeletal events (SSEs) requiring EBRT for pain. In ALSYMPCA, Ra improved overall survival and delayed time to first SSE (Parker NEJM 2013). This post hoc study analyzed ALSYMPCA pts with EBRT for bone pain before randomization and duringtreatment (tx). Methods: Eligible pts had symptomatic CRPC with ≥2 bone mets and no known visceral mets; had best standard of care; and had prior docetaxel (pD) or were unfit for, lacked access to, or declined docetaxel (no pD). Pts were stratified by prior docetaxel use (yes/no), baseline total alkaline phosphatase level (ALP; <220 U/L or ≥220 U/L), and current bisphosphonate use (yes/no), and randomized 2:1 to 6 injections of Ra (50 kBq/kg IV q 4 wk) or placebo (pbo). EBRT use for bone pain was assessed. Adverse events were analyzed by concomitant EBRT. Results: Of 921 ALSYMPCA pts, 30% (186/614) Ra pts and 34% (105/307) pbo pts had EBRT for bone pain. Time to EBRT was significantly longer with Ra vs pbo (HR = 0.67, P = 0.001). Ra vs pbo delayed time to EBRT in pts with <20 mets (HR = 0.49, P < 0.001), current bisphosphonate use

82

Oncol Res Treat 2015;38(suppl 5):1–270

(HR = 0.47, P = 0.004), total ALP < 220 (HR = 0.66, P = 0.008), and no pD (HR = 0.65, P = 0.038). In pts with prior EBRT, a higher percentage of pbo vs Ra pts (33% vs 24%) had EBRT for bone pain during the 6 mo of tx. In pts with no prior EBRT, this percentage was also higher for placebo (33% vs 20%), and the difference was even greater at 12 mo after randomization (41% vs 29%). Ra safety profiles were similar with and without concomitant EBRT. Conclusions: Ra delays the need for EBRT for bone pain vs pbo. Prior EBRT does not appear to affect need for EBRT for bone pain in pts receiving Ra. EBRT use does not affect the Ra favorable safety profile. Disclosure: Petra Strölin: Other Financial Relationships: Support for travel cost for scientific congresses. SI Nilsson: Advisory Role: has had a consultant or advisory relationship with Algeta and has received remuneration from Bayer HealthCare for travel costs and accommodation for study and writing meetings. P265

Effects of radium-223 dichloride (Ra-223) on health-related quality of life (HRQoL) assessed by the EQ-5D utility scores in ALSYMPCA Strölin P.1, Cislo P.2, Sartor O.3, Reuning-Scherer J.4, Shan M.5, Zhan L.2, Parker C.6 Martini-Klinik am UKE GmbH, Hamburg, Germany, 2Bayer HealthCare, Global Health Economics and Outcomes Research Department, Whippany, United States, 3Tulane Cancer Center, Departments of Medicine and Urology, New Orleans, United States, 4Yale University, Department of Statistics, New Haven, United States, 5Bayer HealthCare, Department of Statistics, Whippany, United States, 6The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Academic Urology Unit, Sutton, United Kingdom 1

Background: Ra-223, a first-in-class α-emitter, significantly improved overall survival versus placebo (pbo) (ALSYMPCA) and had a positive impact on pain and HRQoL in patients (pts) with castration-resistant prostate cancer (CRPC) and symptomatic bone metastases (mets) (Nilsson. ASCO GU 2013). In this HRQoL post hoc analysis, EQ-5D-based utility scores were assessed using treatment (tx), disease progression, and symptomatic skeletal events (SSEs) as predictors. Methods: EQ-5D, a preferred measure of HRQoL for economic evaluation by the UK National Institute for Health and Clinical Excellence, was used to determine HRQoL utility scores at baseline, weeks 16 and 24, and 8 follow-up visits using UK tariffs. Repeated measures linear regression models were used to estimate tx effects on utility scores for disease states based on measures of disease progression: prostate-specific antigen (PSA), alkaline phosphatase (ALP), and SSEs. Models were adjusted for baseline utility score and trial stratification factors. Least-squares mean (LSMean) utility scores were estimated for each disease state. The relationship between survival and disease states were described with Kaplan-Meier curves and time to death estimates. Results: LSMean utility score (general measure of HRQoL; score of 1 [full health], 0 [death]) was higher for Ra-223 pts versus pbo pts over the entire trial period and the period prior to an SSE. A tx benefit with Ra-223 was seen in pts with stable disease and prior to an SSE. If a pt experienced ALP progression prior to an SSE, the LSMean utility score decreased (0.62 vs 0.50). However, if progression is defined by PSA, there is little effect of progression on the LSMean utility score (0.61 vs 0.59). There were no tx differences post SSE regardless of ALP or PSA progression. Conclusion: Ra-223 tx benefit in HRQoL is greatest in stable disease/ pre-SSE state. Utility analysis results suggest that ALP and SSE are more appropriate than PSA as measures of progression in pts with CRPC and symptomatic bone mets. Disclosure: Petra Strölin: Other Financial Relationships: Support for travel cost for scientific congresses. Christopher Parker: Advisory Role: has had a consultant or advisory relationship with Algeta, Bayer, and BNIT. He has also received honoraria from Amgen, Astellas, Bayer, Janssen, Sanofi-Aventis, and Takeda.

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

P266

P267

Evaluation of safety, tolerability and activity of Sunitinib in patients (pts) with advanced or metastatic renal cell carcinoma (a/mRCC) in routine clinical practice: the STAR-TOR registry

Evaluation of safety, tolerability and activity of Temsirolimus in patients (pts) with advanced or metastatic renal cell carcinoma (a/mRCC) in routine clinical practice

Woike M.1, Niedtner R.1, Krekeler G.1, Bergmann L.2, Steiner T.3, Goebell P.J.4, Göhler T.5, Harich H.-D.6, Herrmann E.7, Rebmann U.8, Kalanovic D.1

Woike M.1, Strunz A.M.1, Krekeler G.1, Bergmann L.2, Steiner T.3, Goebell P.J.4, Göhler T.5, Harich H.-D.6, Herrmann E.7, Rebmann U.8, Kalanovic D.1 Pfizer Pharma GmbH, Onkologie, Berlin, Germany, 2Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt, Germany, 3Helios-Klinikum, Urologie, Erfurt, Germany, 4AURONTE (Urologische Klinik und Hämatologie/Onkologie (Med 5)), Universitätsklinikum, Erlangen, Germany, 5Onkozentrum, Dresden, Germany, 6Onkologie Hof MVZ GmbH, Hof, Germany, 7Universitätsklinikum Münster, Urologie, Münster, Germany, 8Diakonissenkrankenhaus Dessau, Urologie, Dessau, Germany

1

Pfizer Pharma GmbH, Onkologie, Berlin, Germany, 2Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt, Germany, 3Helios-Klinikum, Urologie, Erfurt, Germany, 4AURONTE (Urologische Klinik und Hämatologie/Onkologie (Med 5)), Universitätsklinikum, Erlangen, Germany, 5Onkozentrum, Dresden, Germany, 6Onkologie Hof MVZ GmbH, Hof, Germany, 7Universitätsklinikum Münster, Urologie, Münster, Germany, 8Diakonissenkrankenhaus Dessau, Urologische Klinik, Dessau, Germany

1

Introduction: Sunitinib (SUN), an oral tyrosine kinase inhibitor, is approved in the EU for the treatment of pts with a/mRCC. A pivotal study had demonstrated significantly increased progression-free survival (PFS) with SUN compared to the former standard interferon alpha (11 vs. 5 months); p < 0.001). To evaluate the safety profile and efficacy of SUN in a clinical routine setting, data collection in a post-approval non-interventional trial seems to be appropriate. Methods: A German multicenter registry for pts with a/mRCC (NCT00700258) was amended to include SUN pts in June 2010 with regulatory and ethic committee´s approval. Objectives are the evaluation of the safety profile of SUN, the tolerability and anti-tumor activity of SUN as well as the profile, comorbidity and characteristics of pts and the sequence of systemic therapies in pts with a/mRCC. Inclusion criteria are histologically confirmed a/mRCC treated with SUN and written informed consent. Results: From July 2010 to February 2015, 108 active study sites recruited 316 pts for this interim analysis. Characteristics: 71.9% male, median age 67.8 years (41.6–87.7), median Karnofsky index 80% (40–100%). Histological subtype: 83.2% clear cell, 16.8% other histologies. In 81.9% of pts SUN is used as first-line therapy. 193 pts were evaluable with regard to MSKCC criteria: 9.3% favorable, 64.2% intermediate, 26.4% poor risk. For all 316 pts, adverse and serious adverse events were observed in 74.7% and 33.9% of the pts (drug related in 60.1% and 14.2% of the pts), respectively. Most common drug-related toxicities (incidence ≥ 5%) of any grade were diarrhea (17.4% of pts), fatigue (13.9%), hand-foot-syndrome (13.0%), thrombocytopenia (11.7%), leukopenia (10.8%), dysgeusia (10.8%), stomatitis (10.4%), nausea (9.8%), anemia (7.6%), mucosal inflammation (7.6%), hypertension (5.7%), and vomiting (5.4%). Median PFS for clearcell a/mRCC pts (n = 263) was 9.0 months (mo), for other histologies 4.2 mo. The subgroup of clear-cell a/mRCC pts for which SUN was first-line therapy (n = 215) had a PFS of 9.2 mo, for second line clear-cell pts (n = 33) 6.7 mo, ≥ third line (n = 14) 11.5 mo . Overall survival (OS) for the entire study cohort was 18.9 mo. Conclusions: The patient population in the registry represents the expected pattern in pts with a/mRCC regarding distribution of age, sex, and histology. Efficacy and tolerability of SUN in routine clinical practice confirms published data.

Introduction: Temsirolimus (TEM), an i.v. mTOR inhibitor, is approved in the EU for the first-line treatment of pts (pts) with a/mRCC who have at least 3 of 6 prognostic risk factors. A pivotal study had demonstrated significantly increased overall survival (OS) with TEM in poor risk pts compared to the former standard interferon alpha (10.9 vs. 7.3 mo; p = 0.0078). To evaluate the safety profile and efficacy of TEM in a clinical routine setting, collection of data in a post-approval non-interventional trial is useful. Methods: A German multicenter registry for pts with a/mRCC treated with TEM was started in January 2008 (NCT00700258) with regulatory and ethic committee´s approval. Objectives are the evaluation of the safety profile of TEM, the tolerability and anti-tumor activity of TEM as well as the profile, comorbidity and characteristics of pts and the sequence of systemic therapies in pts with a/mRCC. Inclusion criteria are histologically confirmed a/mRCC treated with TEM and written informed consent. Results: From February 2008 to February 2015, 108 active study sites recruited 535 pts. Characteristics: 68.9% male, median age 66.9 years (34.9–87.0), median Karnofsky index 80% (40–100%). Histological subtype: 75.3% clear cell, 11.6% papillary, and 2.2% chromophobe RCC. 182 pts were evaluable with regard to modified MSKCC criteria. 78.6% of these pts are classified as poor risk. Median number of metastatic sites is 2 (0–6). Adverse and serious adverse events are observed in 72.7% and 43.4% of the pts (drug related in 41.3% and 9.5% of the pts), respectively. Most common drug-related toxicities (incidence ≥ 3%) of any grade are fatigue (7.9% of pts), anemia (7.3%), rash (6.4%), pruritus (6.1%), oedema of any kind (5.9%), mucosal inflammation (4.1%), thrombocytopenia (3.9%), nausea (3.7%), diarrhoea (3.7%), and stomatitis (3.7%). Median progression-free survival for the total patient population is 4.4 months. A subgroup analysis of pts with baseline LDH ≤ 300 U/l (n = 299) vs > 300 U/l (n = 106) shows significant longer OS for the low LDH group (11.4 vs. 7.2 months, p = 0.015). Median OS for all pts is 10.5 months. Conclusions: The patient population in the registry represents the expected pattern in pts with a/mRCC regarding distribution of age, sex, and histology. Safety and efficacy data of TEM in routine clinical practice confirm current phase III data. Baseline LDH > 300 U/l seems to be a negative prognostic parameter for OS.

Disclosure: Michael Woike: Employment or Leadership Position: Pfizer Pharma GmbH, Deutschland. Daniel Kalanovic: Employment or Leadership Position: Pfizer Pharma GmbH, Deutschland.

Disclosure: Michael Woike: Employment or Leadership Position: Pfizer Pharma Gmbh, Deutschland. Daniel Kalanovic: Employment or Leadership Position: Pfizer Pharma GmbH, Deutschland. P268

Decision making in metastatic renal cell cancer – A retrospective analysis of predictive factors for application of 2nd line therapy Eggers H.1, Ganser A.1, Grünwald V.1 Medizinische Hochschule Hannover, Hannover, Germany

1

Introduction: In renal cell cancer (RCC) not eligible for a curative treatment option tyrosinekinase inhibitors (TKI) have become the standard treatment for first line therapy. Thereby treatment options have grown sig-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

83


CONTENTS AUTHOR INDEX

AUTHOR INDEX

nificantly compared to the era of interleukin-therapy. Consequently new prognostic scores, risk stratification and treatment algorithms had to be developed. Nevertheless we are still in need for information to improve personalized treatment pathways. The aim of this analysis was to determine differences between patients who received a second line therapy to patients who did not (for whatever reason). Material and methods: A random sample of 161 patients treated for RCC between 2005 and 2012 was included. Clinicopathologic data was evaluated Chi-square or Fisher´s exact tests. Continuous parameters were compared applying the Mann-Whitney test. Survival analysis was conducted using Kaplan-Meier analysis, univariate and multivariate cox regression analysis. Results: There was no significant difference between clinical baseline parameters in patients receiving a 2nd line therapy to patients who did not (table 1). In univariate analysis receiving a 2nd line therapy was associated with a significantly better outcome (HR 1,75; p = 0,008; table 2). 3-year overall survival (OS) was 71% in patients with 2nd line compared to 47,6% without (p = 0.007; Fig 1). Progression within 6m of therapy occurred more often in patients receiving no 2nd line therapy (27% vs. 21%, respectively, p = 0.063). Interestingly, the fraction of patients receiving no 2nd line therapy with progressive disease (PD) in less than 6m was larger than the same fraction in patients with PD in more than 6m (40% vs. 21%, p = 0.063). Due to small sample sizes there was no clinicopathologic attribute at all significantly associated with OS in multivariate analysis. Conclusion: The major limitations of our analysis are it’s retrospective nature, small sample size and missing values. A predictive baseline characteristic could not be identified. 2nd line therapy was given irrespective of MSKCC risk category, but early progression was seen more frequently among patients without 2nd line treatment. Improvement for 3-year-OS was detected in patients who were eligible for a 2nd line therapy, underscoring the relevance of continuous treatment in RCC. Disclosure: No conflict of interest disclosed. P269

Marine compound Frondoside A effectively inhibits proliferation of urothelial carcinoma cell lines independent of p53 activity Madanchi R.1, Dyshlovoy S.1,2,3, Honecker F.4, Otte K.1, Alsdorf W.1, Schumacher U.1, Hauschild J.1, Bokemeyer C.1, von Amsberg G.1 UKE, Hamburg, Germany, 2G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Vladivostok, Russian Federation, 3Far Eastern Federal University, Vladivostok, Russian Federation, 4ZeTuP, St. Gallen, Switzerland 1

Introduction: Despite the progress in the therapy of advanced or metastatic urothelial carcinoma, Vinflunin is the only second line treatment approved after failure of Cisplatin based therapy to date – however, with only moderate activity. Thus, novel therapeutic strategies are urgently needed. We examined the efficacy of Frondoside A – a marine natural compound belonging to the group of triterpene glycosides, in a human urothelial carcinoma model. Methods: Anticancer activity of Frondoside A (FronA) was investigated using the 6 human urothelial carcinoma cell lines. The effects on cell viability and cell cycle were investigated using trypan blue staining, MTT cytotoxicity assay, and flow cytometry. Mode of action was analyzed by Western blot analyses. The impact of p53 activity for the cytotoxic activity of FronA was analyzed using siRNA transfection and co-treatment with pifithrin-α – a chemical inhibitor of p53 activity. Synergistic effects of FronA in combination with Cisplatin were investigated using the Chou-Talalay method. Results: FronA significantly inhibited proliferation and reduced viability of 6 human urothelial carcinoma cell lines in a dose dependent manner with IC50 of 0.5~2 µM, while IC50 of Cisplatin were higher ranging from 2~6 µM. The cell lines used for investigation has been the following: RT112, RT-4, HT-1197, TCC(sup), T-24 and 489p. FronA induced apoptosis by caspase-3, -8, and -9 activation, as well as PARP cleavage. Note, inhibition of caspase-3 activity did not suppress the pro-apoptotic activity of the

84

Oncol Res Treat 2015;38(suppl 5):1–270

drug, suggesting caspase-independent character of the induced apoptosis. Inactivation of ERK and p38, and activation of JNK as well as up-regulation of pro-apoptotic Bax were detected. Up-regulation of p21 was observed, although there were no significant effects on cell cycle progression. Silencing of p53 gene via siRNA transfection or inhibition of p53 activity by pifithrin-α did not abolish the cytotoxic effect of FronA. In contrast, Cisplatin acivity was significantly reduced. Furthermore synergistic effects of FronA with Cisplatin were detected in different urothelial carcinoma cell lines. Conclusion: Marine compound Frondoside A is a promising novel therapeutic option for the treatment of advanced or metastatic urothelial carcinoma showing high efficacy in vitro. Inductions of p53- and caspase-3-independent apoptosis are its suggested modes of action. Currently in vivo efficacy and toxicity of FronA are examined. The first and second author contributed equally. Disclosure: No conflict of interest disclosed. P270

A diagnostic and therapeutic challenge – CD 138 positive cells in bone marrow in a patient with urothelial carcinom of the renal pelvis Müller L.1, Tiemann M.2, Schulz H.3, Haferlach T.4 Onkologie UnterEms, Leer-Emden-Papenburg, Leer, Germany, Hämatopathologie, Hamburg, Germany, 3Überregionales Institut für Pathologie, Wilhelmshaven, Germany, 4Münchner Leukämielabor MLL, München, Germany 1 2

Background: Plasmacytoid urothelial carcinoma (PUC) is a rare and aggressive var. bladder cancer (BC) that mimics plasmacytoma cytologically, that can be confused with hemolymphoid neoplasms secondarily affecting the urinary bladder. For the first time to our knowledge we describe a pat. with an urothelial carcinoma (UC) of the renal pelvis who presented with a plasmocytoid CD 138+ UC. Patient: In 01/15 the pat. presented with macrohematuria. In ACT was a left sided renal pelvis mass. In biopsy was a UC G3, nephroureterectomy + lymphadenectomy was performed, no signs of metastatic disease could be detected. The histology showed an UC, G3 left renal pelvis + infiltration in the pararenal adipose tissue, no N+ (0/6). 03/15 anemia, thrombopenia, elevated LDH + diffuse bone pain. In bone scan showed diffuse bone metastasis in skull, total spine, pelvis, ribs, humerus+femura). Retrospectively, preop. CT scans showed no sign for bone metastases. In BM was a diffuse infiltration of plasmocytoid cells up to 80% with suspicion for multiple myeloma with CD 138+ cells. Myeloma could be ruled out by immune cytology + IH with AE1/3, CK20 +CK7 pos. By FISH + selection for CD138+ cells (purity in cytospin >80% of cells) the cells showed trisomie 1 and trisomie 17. A thoroughful reexamination of the primary showed focal PUC Conclusions: We described for the first time to our knowledge a PUC of the renal pelvis. We detected this PUC by chance because of the bone metastases mimicing undifferentiated myeloma cells. Until 2012 only 62 cases of PUC have been described in the literature, but only in the bladder[i] [ii]. All published data of the PUC of the bladder showed all a dismal prognosis, much worth than papillary urothelial carcinoma with early metastases, specially in the focal PUC. In older series the 5-years DFS in T3+ tumors ranged from 8%-40%[iii] [iv]. As in our case the PUC was detected mostly in G3 tumors, sometimes after reexamination and they all were CD 138-positive. In FISH we could describe a trisomie 1 and trisomie 17 in a part of the tumor cells as investigated in the bone marrow after CD138 selection. We think esp. in G3 UC one should investigate esp. for CD138+PUC because of the bad prognosis + develop new therapies.

Abstracts


CONTENTS AUTHOR INDEX

References: 1 Fritsche HM et al.: J Urol., 180(5);2008:1923–1927. 2 Wang Z et al. Int J Clin Exp Pathol. 5;2012:601–608. 3 Ghoneim MA, 180(1):2008:121–127. 4 Shariat SF et al.: J Urol.,176;2006:2414–2422. Disclosure: No conflict of interest disclosed. P271

Efficacy and safety of sequential high-dose chemotherapy (HDCT) for relapsed or refractory germ cell cancer in a unicentric standardized approach Niegisch G.1, Henn A.1, Zaum M.1, Kobbe G.2, Haas R.2, Schirren J.3, Albers P.1, Lorch A.1 Universitätsklinikum Düsseldorf, Urologische Klinik, Düsseldorf, Germany, Universitätsklinikum Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany, 3Horst-Schmidt-Kliniken, Klinik für Thoraxchirurgie, Wiesbaden, Germany 1 2

Introduction: HDCT with carboplatin and etoposide (CE) followed by residual tumor resection (RTR) is a routine regimen for patients (pts) with refractory germ cell cancer (rGCC). We evaluated the results of a unicentric approach with one oncology, one urologic and one thoracic surgeon. Methods: All 26 pts who underwent HDCT for rGCC between 03/12 and 02/2015 were analyzed. Mobilization chemotherapy (paclitaxel/ifosfamide [TI] or VIP) was followed by 3 cycles of high-dose CE (500 mg/m² each, d 1–3). Whenever feasible, all residual tumors were resected immediately after HDCT. Kaplan-Meier method was used to estimate progression-free (PFS) and overall survival (OS). Results: Median age of pts was 41 [range 20–57] years. Histology was non-seminoma in 20/26 pts, 24/26 pts had gonadal primaries. HDCT as 1st salvage was performed in 19/26 pts (prognostic score: 5/19 very low/ low, 9/19 intermediate, 5/19 high/very high risk) and as 2nd salvage in 7/26 pts TI for mobilization was used in 24/26 pts, VIP in 2/26 pts. Median time from mobilization to d1 HDCT was 14 [range 8–27] days. Stem cell harvest was sufficient for 3 cycles in all pts. All HDCT cycles were administered in 22/26 pts. Dose-adjustment due to upfront impaired renal function was required in 4/26 pts. Median time between HDCT cycles was 28 [range 21–36] days. Neither treatment-related deaths nor renal failure requiring hemodialysis was observed. Major toxicities occurred in 5/25 pts (4x sepsis, 1x ileus). Complete response by HDCT alone was achieved in 5/26 pts, 13/26 pts got one or more RTRs. Median time to first RTR was 41 [range 27–72] days. Major surgical complications were observed in 2/13 pts (1x urinoma, 1x wound dehiscence). Histologically 7/13 pts had viable cancer, 2/13 teratoma only and 4/13 necrosis. Maximum response to protocol was CR in 14 pts, PRm- in 7 pts, PRm+ in 4 pts and PD in 1 of 26 pts. Median follow-up was 16.3 [range 4.2–34.3] months with 1-year PFS and 1-year OS of 56 [95% CI 39–79] and 78 [95% CI 62–97]%. Conclusions: A unicentric approach in a high-volume center of expertise provides low complication rates both during HDCT and perioperatively and enables salvage treatment in an optimal timely manner. Even limited by a short follow up outcomes of our series emphasizes the efficacy of HDCT. Disclosure: No conflict of interest disclosed.

P272

Efficacy of kinase inhibitors in germ cell cancer cell lines with differential cisplatin sensitivity Schaffrath J.1, Schmoll H.-J.1, Müller-Tidow C.1, Müller T.1 Martin-Luther-Universität Halle-Wittenberg, Klinik für Innere Medizin IV – Hämatologie und Onkologie, Halle (Saale), Germany 1

Introduction: Testicular germ cell tumors (GCTs) are the most common malignancy in young adult men between the age of 15 and 35. Although cisplatin-based chemotherapy is highly effective even in advanced stages, approximately 20% of patients develop cisplatin resistance which is associated with an unfavorable prognosis. Therefore, new therapeutic options capable of overcoming this resistance are urgently needed. We examined the efficacy of kinase inhibitors targeting mTOR (RAD001), EGFR, HER2/neu, VEGFR (AEE788) and IGF-1R (AEW541) and their influence on cisplatin resistance in germ cell cancer cell lines with differential cisplatin sensitivity. Methods: The in vitro activity of RAD001, AEE788 and AEW541 in the cisplatin sensitive GCT cell lines H12.1 and GCT72F as well as in the resistant cell lines H12.1RA, H12.1ODMneu, 1411HP and 1777NR was investigated using the sulforhodamin-B-(SRB)-cytotoxicity-assay. The agents were applied alone and in combination with cisplatin. To evaluate the activity of the kinase inhibitors, western blot analysis of both the targeted receptors and their phosphorylated state was performed both before and after exposure to each substance. Results: The investigated kinase inhibitors RAD001 (IC50 0,17 to > 10 µM), AEE788 (IC50 0,13 to > 10 µM) and AEW541 (IC50 0,21 to 1,80 µM) showed cytotoxic activity in all GCT cell lines and significantly inhibited proliferation in both cisplatin sensitive and resistant cell lines. While the examined cell lines showed different expression profiles of the targeted receptors, there was no correlation between their expression and phosphorylation and the effectiveness of the agents. The combination of cisplatin and kinase inhibitors showed both additive and antagonistic effects in the studied cell lines, yet no significant synergistic effect was observed. Conclusion: According to our data, the investigated kinase inhibitors are effective in germ cell cancer cell lines independent of their cisplatin sensitivity. However, when combined with cisplatin they do not demonstrate any promising ability to overcome cisplatin resistance in testicular GCTs. Disclosure: No conflict of interest disclosed. P273

β-1,4-Galactosyltransferase-I expression in peripheral T-lymphocytes is associated with relapse-free survival in testicular cancer patients Nilius V.1, Timmesfeld N.2, Neubauer A.1, Brendel C.1 Uni Marburg, Hämatologie, Marburg, Germany, 2Institut für medizinische Biometrie und Epidemiologie, Marburg, Germany 1

Outcome of germ cell tumor patients with progression or relapse after a cisplatin-based first line chemotherapy is highly heterogeneous, ranging from 10 to 70% overall survival. As growing evidence suggests that immunosurveillance is important for tumor control we investigated β-1,4-Galactosyltransferase-I (B4GALT1) expression levels in a cohort of 49 testicular cancer patients. High expression levels of B4GALT1 in peripheral blood T-lymphocytes were associated with a better relapse-free survival (RFS) (p ≤ 0.006). B4GALT1 is a cell surface molecule involved in immune cell cross-talk, expression levels proved to be more accurate in predicting RFS than the IGCCCG scoring system in our patient cohort (p ≤ 0.006 versus 0.068). Moreover, interleukin 10 (IL10), a cytokine released by cytotoxic T-cells, was also significantly elevated in T-lymphocytes of non-relapse germ cell tumor patients (p ≤ 0.025). Our data indicate that T-lymphocyte biology may play a pivotal role in disease control in testicular cancer patients with relapsed or progressive disease state. Disclosure: No conflict of interest disclosed.

86

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Fortbildung

Disclosure: Uwe Platzbecker: Financing of Scientific Research: Celgene, Novartis, Amgen; Expert Testimony: Celgene, Novartis, Amgen.

V274

Wissenschaftliches Symposium

MDS Differentialdiagnose und Therapiemodalitäten

MDS: Diagnosis and differential diagnoses

NHL indolent neue Substanzen

Pfeilstöcker M.1 Hanusch KH, 3. Med Abt, Wien, Austria

1

Despite recent developments in diagnostic tools and methods diagnosis and differential diagnoses are still a challenge in myelodysplastic syndromes (MDS). One reason is the heterogeneity of the disorder with many different underlying molecular mechanisms, therefore there is still no single test available that defines MDS unambiguously. In addition the slow onset of MDS especially in low risk cases poses diagnostic difficulties in early disease. In principle each cytopenia that cannot be explained by other reasons such as vitamin deficiencies, infections, autoimmune or metabolic disorders or toxic effects on the bone marrow has to be evaluated for the presence of MDS, while other clonal hematopoietic disorders such as leukemias, paroxysmal nocturnal hemoglobinuria, aplastic anemia, myeloproliferative neoplasms (myelofibrosis) or lymphoma (hairy ell leukemia) have to be considered as well. Diagnosis of MDS is made by bone marrow puncture (cytology including iron staining and histology). After exclusion of other reasons evidence of dysplasia according to published diagnostic criteria establishes the diagnosis. Metaphase cytogenetics complemented by FISH analysis is mandatory also for prognostication, FACS and molecular analyses are still optional as their role in classification and prognosis is presently being established. IDUS (idiopathic dysplasia of undetermined significance) and ICUS (idiopathic cytopenia of undetermined significance) designate states where the diagnostic criteria of MDS are not yet fulfilled, recently a partially overlapping state of CHIP (clonal hematopoiesis of indeterminate potential) – with ascertained clonal hematopoiesis but not necessarily cytopenia – has been described with yet unclear clinical consequences. After a diagnosis of MDS has been made subtypes according to the WHO classification should be defined and prognostic risk groups assessed using instruments such as the recently improved international prognostic scoring system IPSS (-R) which uses number and degree of cytopenias, bone marrow blast count and refined cytogenetics as prognostic parameters that may be complemented with differentiating features such as age. Assessment of ferritin, endogeneous erythropoietin levels and optionally HLA typing completes initial work up. Combining these disease related features with patients´ resources (performance status, comorbidities, geriatric assessment) finally provides the tools for clinical decision making. Disclosure: No conflict of interest disclosed. V276

Therapy in MDS Platzbecker U.1, Deutsche MDS SG Uniklinik, Dresden, Germany

1

The diversity of MDS makes the disease challenging and “truly personalized” not only in terms of diagnostics but also in carrying out clinical decisions. The heterogeneity of MDS manifests in the individual patient as a disease ranging from an indolent condition with a considerable life expectancy to forms approaching the aggressiveness of acute myeloid leukemia (AML). A risk-adapted treatment strategy is therefore mandatory for a disease showing such a highly variable clinical course. Prognostic factors may be subdivided into those related to the general patient’s characteristics and health condition and those related to the MDS disease itself. During the past decades treatment has been stratified according to the International Prognostic Scoring System (IPSS) risk score; i.e. into “lower-risk” MDS (low/int-1, LR-MDS) where correction of cytopenia was the main objective and “higher-risk” MDS (int-2/high, HR-MDS) where the reduction or delay of progression or AML evolution and prolonged survival was the objective.

Abstracts

V277

Antibody-drug conjugates in indolent lymphoma Viardot A.1 Klinik für Innere Medizin III, Universitätsklinikum, Ulm, Germany

1

Monoclonal antibodies revolutionized many fields of hemato-oncology. Antibody-drug conjugates (ADCs) realize ideally the Paul Ehrlich´s conception of “magic bullets”. Recently, two ADCs were approved by the European Medical Agency (EMA): brentuximab vedotin in Hodgkin´s disease and anaplastic large cell lymphoma and trastuzumab emantansine in HER2+ metastatic breast cancer. In follicular lymphoma (FL), there is a long-term experience with radioimmunoconjugates (RIC). For example, Yttrium-90 Ibritumomab is approved in relapsed FL and as consolidation therapy after chemoimmunotherapy. In contrast to RICs, the ideal target for ADCs is internalized after binding of the antibody. CD20, which remains stable at the cell surface after binding, is less suitable than the targets CD19, CD22, CD37, CD74 or CD79b. The most frequently used toxins – aurastatins, calicheamicins and maytansinoids – are synthetically derived from natural compounds and too toxic for direct application. Few drugs have already finished phase-I/II studies including patients with indolent lymphomas. Inotuzumab ozogamicin (anti-CD22) has an overall response rate (OR) of 68% in FL at maximum tolerated dose (MTD) (Advani A et al., J Clin Oncol 28:2085, 2010). Polatuzumab vedotin (against CD79b) shows an OR 47% in FL at MTD (MC Palanca-Wessels et al., Lancet Oncology 2045:70128, 2015). Coltuximab-ravtansine (against CD19) has a response rate of 40% in FL (V Ribrag, Clin Cancer Res 20:213, 2014). Addition to Rituximab increased the response rate in inotuzumab ozogamicin (OR 87% in FL, L Fayad et al., J Clin Oncol 31:573, 2013) and polatuzumab vedotin (OR 60% in FL, F Morschhauser et al., J Clin Oncol 32:5s, 2014 (suppl; abstr 8519)). However, the first randomized trial with inotuzumab ozogamicin and rituximab versus the investigator´s choice was terminated due to slow accrual (NCT562965). New promising combinations of ADCs with other drugs will be tested in ongoing clinical trials. Antibody-drug conjugates will complement the therapeutic options in indolent lymphomas in the next years. Disclosure: Andreas Viardot: Advisory Role: Janssen, Gilead, Amgen, Roche; Financing of Scientific Research: Janssen, Roche, Pfizer; Other Financial Relationships: Reisekosten: Roche, Amgen. V279

B-cell receptor signaling in lymphoid malignancies – oncogenic driver and therapeutic target Schmitt C.1 Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany 1

B-cell receptor (BCR) signaling emerged as a key therapeutic target in lymphoma. In normal B-cells, the BCR determines antigen specificity and marks their unique identity. Composed of two identical heavy and light chains, the BCR is the product of a complex B-cell-specific genomic affinity maturation process that includes V(D)J recombination, somatic hypermutation and class switch recombination. Typically following ligand-induced receptor aggregation, the BCR mediates phosphorylation of the associated Ig-a/Ig-b (CD79A/CD79B) immunoreceptor tyrosine-based activation motifs (ITAMs) by SRC kinases, which initiates a cascade of downstream events including the Bruton’s Tyrosine Kinase (BTK), phosphatidylinositol 3-kinase (PI3K) and NF-kB signaling, collectively amplifying the original BCR signal. Antigen-dependent and potentially

Oncol Res Treat 2015;38(suppl 5):1–270

87


CONTENTS AUTHOR INDEX

ligand-independent “tonic” BCR signaling is essential for normal B-cell survival and function. Importantly, most mature B-cell malignancies rely on functional BCR signaling as well, although the underlying mode of activation may be diverse (including tonic signaling, exposure to [auto]-antigens, or self-aggregation of the BCR). This is further underscored by their usage of a restricted, non-random set of stereotyped IgH V segments, aberrant somatic hypermutation, and activating mutations in signaling components (such as CD79/ITAM or CARD11) of the BCR cascade. B-cell lymphomas may present with BCR signaling-related vulnerabilities that can be exploited by novel targeting approaches. Most prominently, the BTK inhibitor Ibrutinib or the d-isoform-specific PI3K inhibitor Idelalisib have been clinically approved, and numerous novel compounds are in pre-clinical or early clinical testing. However, functionally poorly understood BCR-governed signaling networks and their different wiring across B-cell lymphoma entities, as well as considerable genetic heterogeneity within histological or even molecularly defined subtypes seem to account for the remarkable diversity in clinical responsiveness to these novel agents. While BCR signaling-targeting agents undoubtedly opened a new era in lymphoma therapy, their specific mode(s) of action and the signaling conditions under which they exert their activity require further investigation regarding enhanced efficacy, less toxicity, personalized stratification and treatment optimization, at least in part, via rational co-targeting of critical collateral pathways. Disclosure: No conflict of interest disclosed.

Freier Vortrag

conjunction with chemotherapy in newly diagnosed elderly Ph+ ALL pts (registered under EudraCT no.:2010–022855–46). Methods: Male or female pts. >55 years with untreated Ph+ and/or BCRABL1 positive ALL were treated after a pre-phase, with nilotinib 400 mg BID during the induction period in combination with weekly vincristine (VCR) and dexamethasone (DEX) for 4 weeks. Consolidation consisted in nilotinib with methotrexate and asparaginase for cycles 1, 3 and 5 and cytarabine for cycles 2, 4 and 6. Maintenance phase consisted of nilotinib sequentially with 6-MP and methotrexate orally one every other month and Dex/VCR every two months up to 24 months of treatment. Results: Between 2012 and 2014, 56 pts. (25 male, 31 female) have been enrolled. Median age is 65 years (55–85 years), twelve pts. are older than 70 years of age. To date, all pts. are evaluable for safety and 47 pts. are evaluable for efficacy. The CHR rate is 87%, one pt. was refractory (2%), one pt. had a partial remission (2%). One pt. died during induction therapy (2%), three pts. discontinued therapy before CR evaluation (6%). With a median follow-up of 5.5 months, 34 of the 41 pts. who achieved CR are in CCR and 3 pts. relapsed, two of whom had discontinued study treatment to undergo allogeneic SCT. 9 pts. have discontinued study treatment prematurely because of transfer to allogeneic SCT, as explicitly permitted by the protocol. The rate of complete molecular remission (B/A < 10E03) after induction (25 pts. evaluable) was 45.5%, 5 pts. had undetectable BCR-ABL1 transcripts. Tolerability has been acceptable. Infectious events and neutropenic fever predominated. Conclusion: Nilotinib in conjunction with chemotherapy according to the EWALL-PH-02 protocol is highly effective and well tolerated in elderly pts. with newly diagnosed Ph+ ALL. Molecular response rates are high and MRD levels in responding pts. decrease with time. Disclosure: Heike Pfeifer: No conflict of interest disclosed. Oliver Ottmann: Advisory Role: Novartis, BMS, Ariad, Pfizer; Financing of ­Scientific Research: Novartis, BMS, Ariad, Pfizer.

ALL V285

European working group for adult ALL phase II trial of nilotinib in combination with chemotherapy for first-line treatment in elderly patients with de novo Philadelphiachromosome positive acute lymphoblastic leukemia (EWALL-PH-02) Pfeifer H.1, Cayuela J.-M.2, Spiekermann K.3, Beck J.4, Jung W.5, Viardot A.6, Schäfer-Eckhart K.7, Reichle A.8, Maury S.9, Schmitz N.10, Heidenreich D.11, Panse J.12, Junghanß C.13, Raffoux E.14, Suarez F.15, Guillerm G.16, Alexis M.17, Lissandre S.18, Huguet F.19, Isnard F.20, Lepetre S.21, Escofffre-Barbe M.22, Ribera J.-M.23, Goekbuget N.1, Dombret H.14, Hoelzer D.24, Rousselot P.25, Ottmann O.1 Goethe University Hospital, Frankfurt/Main, Medizinische Klinik II, Frankfurt am Main, Germany, 2Hôpital Saint Louis, Molecular Biology Department, Paris, France, 3Ludwig-Maximilians-Universität, München, Germany, 4 Universitätsklinik, Mainz, Germany, 5Universitätsklinik, Göttingen, Germany, 6 Universitätsklinik, Ulm, Germany, 7Klinikum Nürnberg Nord, Nürnberg, Germany, 8Universitätsklinik, Regensburg, Germany, 9Hopital Henri Mondor, Creteil, France, 10Asklepios Klinik St. Georg, Hamburg, Germany, 11 Universitätsmedizin, Mannheim, Germany, 12RWTH Aachen University Hospital, Aachen, Germany, 13Universitätsklinik, Rostock, Germany, 14Hopital Saint-Louis, APHP, Paris, France, 15Hopital Necker, Paris, France, 16Hopital Norvan, Brest, France, 17CHR Orleans, Orleans, France, 18CHU de Tours, Tours, France, 19Hopital Purpan, University, Toulouse, France, 20Hopital Saint Antoine, APHP, Paris, France, 21CLCC H Becquerel, Rouen, France, 22CHU Rennes, Rennes, France, 23Universitat Autonoma de Barcelona, Barcelona, Spain, 24Onkologikum, Frankfurt am Main, Germany, 25Universite Versailles Saint-Quentin-en-Yvelines, Versailles, France 1

Introduction: Imatinib in combination with chemotherapy is considered to be the gold standard for treatment in patients with Philadelphia positive (Ph+) acute lymphoblastic leukaemia (ALL). But the overall prognosis particularly in elderly patients (pts.) remains poor. Nilotinib is a more effective second generation ABL kinase inhibitor (TKI). Data on its efficacy in Ph+ ALL is limited. The EWALL (European Working Group for Adult ALL) initiated a prospective, investigator-initiated multicenter European clinical trial to examine the efficacy and safety of nilotinib in

88

Oncol Res Treat 2015;38(suppl 5):1–270

V286

Silencing of GATA3 defines a new stem cell subgroup of T-ALL Fransecky L.1, Neumann M.1, Heesch S.1, Schlee C.1, Ortiz Tanchez J.1, Heller S.1, Schwartz S.1, Mochmann L.H.1, Isaakidis K.1, Bastian L.1, Mossner M.2, Herold T.3, Goekbuget N.4, Baldus C.D.1 Charité, University Hospital Berlin, Campus Benjamin Franklin, Hematology/ Oncology, Berlin, Germany, 2University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Hematology/Oncology, Mannheim, Germany, 3University Hospital Grosshadern, Ludwig-Maximilians-University (LMU), Department of Internal Medicine III, Munich, Germany, 4Goethe University Hospital, Hematology/Oncology, Frankfurt/Main, Germany 1

Introduction: The critical role of GATA3 is well characterized in later stages of T-cell differentiation, but its role for the generation of early-Tcell precursors (ETP) has only recently been explored. As aberrant GATA3 mRNA expression has been linked to tumorigenesis, we here investigated the role of GATA3 in T-cell acute lymphoblastic leukemia (T-ALL). Patients and methods: Seventy-one ETP-ALL samples, sent to the central diagnostic reference laboratory of the German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia, were investigated by RT-PCR analysis for GATA3 mRNA expression. In addition, pyrosequencing was performed to determine GATA3 DNA methylation in 69 of 71 samples. Additionally, global DNA methylation was assessed in 12 ETP-ALL samples using the Illumina Infinium® HumanMethylation450 BeadChip platform. Microarray expression data (Affymetrix HG-U133 Plus 2.0) of an independent cohort of 83 T-ALL patients, which included both ETP-ALL and non-ETP-ALL patients, was used for read out of a GATA3-specific gene expression profile. Results: One third of ETP-ALL samples lacked GATA3 expression (23/71, 32%, GATA3null ETP-ALL). Global DNA methylation analysis revealed a 6kb segment within the GATA3 locus with significant DNA hypermethylation (46% v. 19%, p < 0.001) containing 35 differentially methylated sites. We confirmed GATA3 hypermethylation in the ETP-ALL cohort (n = 69) and found that GATA3 mRNA expression and DNA methylation were inversely correlated (r = -0.73, p < 0.001). GATA3null ETP-ALL was char-

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

acterized by a high frequency of FLT3 mutations (15/19, 79%) and a low frequency of NOTCH1 mutations (2/19, 11%). Gene set enrichment analysis of GATA3null in an independent cohort of T-ALL revealed enrichment of genes associated with hematopoietic stem cells and depletion of genes involved in T cell differentiation. Among upregulated genes in GATA3null samples, we identified molecules such as FLT3, C-KIT, BCL2, and BTK that could potentially be accessible for targeted therapies. Conclusion: GATA3 silencing occurs in a third of adult ETP-ALL patients and is associated with GATA3 DNA hypermethylation. While GATA3positive ETP-ALL might be susceptible to conventional lymphoid directed therapies, GATA3null ETP-ALL exhibits stem cell leukemia properties, which might be associated with distinct sensitivities towards targeted therapies. Disclosure: No conflict of interest disclosed. V287

Droplet digital PCR to quantify minimal residual disease in acute lymphoblastic leukemia Koopmann J.1, Pott C.1, Füllgrabe M.1, Knecht H.1, Gökbuget N.2, Kneba M.1, Brüggemann M.1 Universitätsklinikum Schleswig-Holstein, Campus Kiel, Medizinische Klinik II, Kiel, Germany, 2Goethe University Hospital, Medizinische Klinik II, Frankfurt, Germany 1

Introduction: Minimal residual disease (MRD) has been proven to be the most powerful prognostic factor in ALL. The current molecular gold standard for MRD quantification is the allele specific real-time quantitative PCR (ASO RQ-PCR) of clonal immunoglobuline (IG) and T-cell receptor (TR) gene rearrangements. However, new high throughput tools emerge to overcome some limitation of RQ-PCR. Therefore, we compared the performance of droplet digital (dd)PCR and RQ-PCR to quantify MRD in ALL. Material and methods: We adapted ASO ddPCR for MRD monitoring in ALL and checked linearity, reproducibility, repeatability, lower limit of detection and lower limit of quantification. Then we analyzed a total of 154 samples (17 diagnostic, 93 follow-up, 44 serial dilutions of diagnostic samples) of adult patients with ALL using IG/TR ASO ddPCR and compared results with standardized RQ-PCR. RQ PCR was performed according to EuroMRD guidelines. Results: Sensitivity and lower limit of quantification of ddPCR as defined by EuroMRD guidelines was comparable to RQ-PCR using the same amount of input DNA. ddPCR results proved to be highly reproducible, even at the lower limit of quantification. Comparison of MRD analysis using ddPCR and ASO RQ-PCR showed good concordance (r = 0.83, p < 0.0001) between both methods. In contrast to RQ-PCR MRD quantification of follow up samples using ddPCR was possible without the usage of serial dilutions of the diagnostic sample as external calibrator thereby significantly reducing labor intensiveness compared to RQ-PCR. In addition, it allowed the exact quantification of the clonal marker in the diagnostic sample. This represents a major advantage of ddPCR compared to RQ-PCR as it enables the exact quantification of tumor load and thereby the identification of oligoclonality at diagnosis even in patients with low infiltrated diagnostic samples. This was highlighted by comparison of the two molecular MRD approaches in one patient where ddPCR of the diagnostic sample identified subclonality of the marker IGH gene rearrangement. In contrast, RQ-PCR of serial dilutions of the calibrating diagnostic sample showed late amplification and poor sensitivity which precluded correct RQ-PCR based MRD quantification of follow-up samples in this case. Conclusion: We showed that ddPCR is a promising tool for IG/TR based MRD analysis in ALL. Prospective analyses of unselected cases have to be performed to verify the clinical impact of ddPCR-based MRD detection. Disclosure: No conflict of interest disclosed.

90

Oncol Res Treat 2015;38(suppl 5):1–270

V288

Treatment of B-lymphoblastic leukemia cells with demethylating agents enhances the sensitivity to cytostatic drugs Konkolefski C.1, Roolf C.1, Sklarz L.-M.1, Sekora A.1, Murua Escobar H.1, Junghanss C.1 Universitätsmedizin Rostock, Klinik III, Hämatologie, Onkologie, Palliativmedizin, Rostock, Germany 1

Introduction: Aberrant methylation of tumorsuppressor gene promoters is frequently observed in acute lymphoblastic leukemia (ALL). Several studies have shown that hypermethylation is an independent prognostic factor for disease-free survival and play a critical role in drug resistance. Azacitidine (AZA) and decitabine (DAC) are methyltransferase inhibitors which partially reverse aberrant DNA methylation and sensitize malignant myeloid cells to cytostatics. However, it is not yet clear whether these agents are equally effective in ALL cells. Therefore, we examined the impact of AZA and DAC on B-ALL cells in single application and in combination with other cytostatics to investigate the sensitizing effect. Methods: B-ALL cell lines SEM and RS4;11 (both harbouring a t(4;11)) were treated with increasing concentrations of AZA or DAC (100–1.000 nM) for up to 72 h. Cell proliferation and metabolism were examined by trypan blue staining and WST-1 testing. Cell cycle and apoptosis changes were analyzed by flow cytometry. For drug combination experiments, cells were exposed to AZA or DAC (500 or 100 nM) in combination with IC80 values of ara-C, dexamethasone and doxorubicin. Thereby, the demethylating agents were applied in different preparations: simultaneously as well as 24 hours before or after the respective cytostatics. Changes in cell proliferation were determined by WST-1 and drug synergy was evaluated using BLISS statistic. Results: In SEM cells, AZA and DAC induced dose-dependent antiproliferative effects starting at a concentration of 100 nM. Metabolic activity was significantly decreased by AZA: to 65.6 ± 15.3% and DAC: to 32.7 ± 9.3% compared to control cells (100%). Both drugs increased the number of cells in G0/G1 stage of the cell cycle and induced apoptosis. Treatment with AZA or DAC was less effective in RS4;11 cells compared to SEM. Enhanced antiproliferative effects on cells were detected when DAC was combined with doxorubicin or ara-C. Here, metabolic activity decreased significantly when DAC and doxorubicin or ara-C were given simultaneously or when cells were pre-treated with doxorubicin, compared to single drug treatment. Conclusion: Demethylating agents possess antiproliferative effects on B-ALL cells in vitro and may be beneficial for combined chemotherapy in ALL. Different response rates of the cell lines on AZA and DAC may be referred to altered methylation patterns, which is currently under investigation. Disclosure: No conflict of interest disclosed. V289

Deletions of CDKN2A/B and PAX5 are frequently restricted to the CD19+ compartment and associated with concurrent deletions of ABL in adult BCR-ABL+ BCP-ALL Bartels M.1, Nagel I.2, Ussat S.3, Ottmann O.4, Pfeifer H.4, Trautmann H.1, Böttcher S.1, Gökbuget N.4, Kneba M.1, Oberg H.-H.3, Siebert R.2, Brüggemann M.1 Universitätsklinikum Schleswig-Holstein, Campus Kiel, 2. Medizinische Klinik, Kiel, Germany, 2Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institut für Humangenetik, Kiel, Germany, 3Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institut für Immunologie, Kiel, Germany, 4Universitätsklinikum Frankfurt, 2. Medizinische Klinik, Frankfurt, Germany 1

Introduction: BCR-ABL fusion positive (BCR-ABL+) B-cell precursor acute lymphoblastic leukemia (BCP-ALL) may originate at distinct stages of hematopoiesis. It is unclear, weather concurrent CDKN2A/B- and PAX5-Deletions can also be assigned to certain hematopoietic compart-

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

ments. Therefore we traced the distribution of driver mutations within relevant hematopoietic compartments of BCR-ABL+ adult BCP-ALL cases with concurrent CDKN2A/B- and PAX5-Deletions in order to detect targetable immunophenotypic patterns of involvement. Methods: Cryo-conserved samples from initial diagnosis of 12 adult ­BCR-ABL+ BCP-ALL patients with known CDKN2A/B deletions treated within the GMALL 07/2003 trial were flow sorted and screened for ­BCR-ABL, CDKN2A/B and in 5 cases also for PAX5 deletions by fluorescence in situ hybridization (FISH) using one customized (PAX5) and two commercial probes (Vysis LSI BCR/ABL Dual Color, Dual Fusion Translocation probe, Vysis LSI CDKN2A/CEP 9; both Abbott Molecular, Illinois, USA). The following compartments were analyzed: CD34+38–19–10–3-, CD34+38+19–3-, CD34+19+20–3-, CD34+19+20+3-, CD34–19+20+3-, CD34–19–20–3+, CD34+19–13/33+10–3/16-, CD34–19–13/33+10– 3/16-, CD34–19–13/33+10–3/16+. Results: Diverse individual subclones with CDKN2A/B deletions were exclusively detectable within the B-committed progenitor compartment (CD34+19+) in 10 out of 12 cases although concurrent BCR-ABL+ multipotent progenitors were evident in 4 cases. Cells with a PAX5 deletion had a CD34+19+ phenotype in all 5 investigated cases, including 3 cases with evident BCR-ABL+ multipotent progenitors. Finally, 10 patients of the cohort showed major leukemic (sub)clones with atypical signal patterns after FISH with the BCR/ABL probe: in 7 cases the major subclone was characterized by a missing ABL signal. Conclusions: While the t(9;22) frequently represents an antecedent event prior to B-lineage determination in the genesis of BCR-ABL+ BCP-ALL this does not hold true for CDKN2A/B and PAX5. In our cohort the different subclones with deletions of CDKN2A/B and PAX5 were commonly restricted to the CD34+19+ B-committed progenitor compartment regardless of BCR/ABL+ multipotent progenitors. Our observation may help to identify patients that particularly profit from B-cell specific immunotherapy. Monosomy 9 probably accounts for the association of ABL-deletions with CDKN2A/B cna in some but not all cases.

overall survival (OS). Logistic regression was used to identify predictors of such modification in dosage. Results: 681 of 690 patients were treated with at least one dose of FCR. Of these, 253 (37.2%) received FCR with either FC, R, or both reduced by 20% or more. These patients had significantly shorter PFS (41.6 vs. 64.0 months, HR = 1.78, 95%CI = 1.45–2.23) and OS (90.2 months vs. not reached, HR = 2.15, 95%CI = 1.57–2.96) compared to patients with a ≤20% reduction in dose (Figure 1–2). In univariate and multivariate analyses, age >60 years, Binet stage C and serum β2-microglobuline >3.5 mg/l were identified as independent predictors for reducing the prescribed dose of FCR. Conclusions: This pooled analysis of two GCLLSG trials showed that reduction of full-dose FCR treatment significantly mitigates its efficacy and results not just in impaired PFS, but also OS. Predictors of dose reductions during therapy with FCR may help to refine treatment decision-making in CLL.

Disclosure: No conflict of interest disclosed.

Disclosure: Gabor Kovacs: Other Financial Relationships: travel grant by ­Mundipharma. Valentin Goede: Advisory Role: F. Hoffmann-La Roche; Financing of Scientific Research: F. Hoffmann-La Roche, Mundipharma, Glaxo Smith Kline, Bristol Myers Squibb.

Freier Vortrag CLL klinisch

Fig. 1

V290

Impact and predictors of reducing prescribed doses of fludarabine, cyclophosphamide and rituximab (FCR) in frontline treatment of chronic lymphocytic leukemia (CLL) Kovacs G.1, Bahlo J.1, Kluth S.1, Fink A.M.1, Cramer P.1, von Tresckow J.1, Maurer C.1, Langerbeins P.1, Groß-Opphoff-Müller C.1, Fischer K.1, Wendtner C.-M.2, Kreuzer K.-A.1, Stilgenbauer S.3, Hallek M.1, Eichhorst B.1, Goede V.1 Uniklinik Köln, Klinik I für Innere Medizin (DCLLSG), Köln, Germany, 2Klinikum Schwabing, Klinik für Hämatologie, Onkologie, Immunologie, Palliativmedizin, Infektiologie und Tropenmedizin, München, Germany, 3Uniklinik Ulm, Klinik III für Innere Medizin, Ulm, Germany 1

Introduction: Frontline treatment with full-dose FCR is considered standard of care for physically fit patients with CLL. In routine practice, however, adverse events often lead to reduction in dose of FCR which might result in a decrease of efficacy. The aim of our study was to systematically evaluate the impact of reducing the prescribed dose of FCR on its efficacy and to identify clinical and biological predictors for such dose modification. Methods: Patients treated with FCR within two randomized phase III trials of the German CLL Study Group (GCLLSG) (CLL8: FC (n = 409) vs. FCR (n = 408); CLL10: FCR (n = 282) vs. bendamustine plus R (n = 279)) were pooled (n = 690). The planned FCR dose (according to protocol) was compared with the actually applied dose in each patient and differences were provided in%. Patients with ≤20% and with >20% reduction in planned dose were compared with regard to progression free (PFS) and

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

91


CONTENTS AUTHOR INDEX

AUTHOR INDEX

V291

V292

Results from the phase 2 RESONATE™-17 trial: Efficacy and safety of Ibrutinib in patients with relapsed or refractory chronic lymphocytic leukemia or small lymphocytic lymphoma with Del17p

Bendamustine and Rituximab in combination with Lenalidomide in patients with relapsed or refractory and in patients with previously untreated chronic lymphocytic leukemia: A multicenter phase I/II safety and efficacy trial of the German Chronic Lymphocytic Leukemia Study Group (GCLLSG)

Stilgenbauer S.1, Jones J.A.2, Coutre S.E.3, Mato A.R.4, Hillmen P.5, Tam C.6, Osterborg A.7, Siddiqi T.8, Thirman M.J.9, Furman R.R.10, Ilhan O.11, Keating M.12, Call T.G.13, Brown J.R.14, Stevens-Brogan M.15, Li Y.15, Clow F.15, James D.F.15, Chu A.D.15, Hallek M.16, O’Brien S.12 University of Ulm, Ulm, Germany, 2The Ohio State University, Division of Hematology, Columbus, United States, 3Stanford University School of Medicine, Division of Hematology, Stanford Cancer Center, Stanford, United States, 4 Hackensack University Medical Center, Hackensack, United States, 5The Leeds Teaching Hospitals, St. James Institute of Oncoogy, Leeds, United Kingdom, 6 Peter MacCallum Cancer Centre, East Melbourne, Australia, 7Karolinska University Hospital Solna, Department of Hematology, Solna, Sweden, 8City of Hope National Medical Center, Department of Hematology/Hematopoietic Cell Transplantation, Duarte, United States, 9The University of Chicago Medicine, Section of Hematology/Oncology, Chicago, United States, 10Weill Cornell Medical College, Division of Hematology-Oncology, New York, United States, 11 Ankara University Medical Faculty, Ankara, Turkey, 12The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, United States, 13 Mayo Clinic, Division of Hematology, Rochester, United States, 14Dana Farber Cancer Institute, Division of Hematologic Malignancies, Boston, United States, 15 Pharmacyclics, Inc., Sunnyvale, United States, 16University Hospital Cologne, Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn, Cologne, Germany 1

Introduction: Patients with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL) with del17p have poor treatment outcomes. Ibrutinib (Imbruvica®) is a first-in-class BTK inhibitor approved by EMA for adult CLL patients with ≥1 prior therapy and first-line CLL patients with del17p or TP53 mutation who are unsuitable for chemotherapy. We report a primary analysis of the Phase 2 RESONATE™-17 trial evaluating efficacy and safety of single-agent ibrutinib in R/R del17p CLL or small lymphocytic lymphoma (SLL). Methods: Patients with del17p CLL/SLL who failed ≥1 prior therapy received oral 420 mg ibrutinib once daily until disease progression. Primary endpoint was overall response rate (ORR) per independent review committee (IRC). Other endpoints were duration of response (DOR), progression-free survival (PFS), overall survival (OS) and safety. The analysis was conducted 12 months after final enrollment and included all patients receiving ≥1 dose of ibrutinib. Results: Of 144 patients, 137 had CLL and 7 had SLL. Median age was 64. At baseline, 63% were Rai Stage III/IV; 49% had bulky lymphadenopathy ≥5 cm. Patients had a median of 2 prior therapies. Investigator-assessed ORR was 83%, with complete response (CR) or CR with incomplete bone marrow recovery (CRi) in 2% of patients and partial response with lymphocytosis (PR-L) in 17%. ORR by IRC was 65%, including 4% PR-L. At 11.5 months of median follow-up, median PFS, OS and DOR had not been reached. The 12-month PFS rate was 79.3% and 12-month OS was 83.5%. Of 20 patients (13.9%) who progressed, 11 (7.6%) had Richter’s transformation. Most common adverse events (AE) were diarrhea (36%), fatigue (31%), cough (24%) and arthralgia (22%); most were Grade 1–2. Most common Grade 3–4 AEs were neutropenia (14%), anemia (8%), pneumonia (8%) and hypertension (8%). Atrial fibrillation occurred in 11 patients (7.6%; 3.5% Grade 3/4) and major hemorrhage in 7 patients (4.9%, all Grade 2/3). Sixteen patients (11.1%) discontinued treatment due to AEs. At data cut, 70% of patients continued on ibrutinib. Conclusions: In the largest prospective study to date of patients with del17p CLL/SLL, ibrutinib demonstrated marked efficacy in measures of ORR, DOR, PFS and OS, with an AE profile similar to that of earlier studies. These results support ibrutinib as an effective treatment for patients with del17p CLL/SLL. Disclosure: Stephan Stilgenbauer: Advisory Role: Pharmacyclics; Financing of Scientific Research: Pharmacyclics. Susan O´Brien: Financing of Scientific Research: Janssen; Expert Testimony: Pharmacyclics.

92

Oncol Res Treat 2015;38(suppl 5):1–270

Pflug N.1, Maurer C.1, Bahlo J.1, Kluth S.1, Rhein C.1, Cramer P.1, Gross-Ophoff C.1, Langerbeins P.1, Fink A.-M.1, Eichhorst B.1, Kreuzer K.-A.1, Tausch E.2, Stilgenbauer S.2, Böttcher S.3, Döhner H.2, Kneba M.3, Hallek M.1, Wendtner C.-M.4, Bergmann M.4, Fischer K.1, Deutsche CLL Studiengruppe Klinik I für Innere Medizin, Uniklinik Köln, Köln, Germany, 2Klinik III für Innere Medizin, Uniklinik Ulm, Ulm, Germany, 3Klinik II für Innere Medizin, Uniklinik Schleswig-Holstein, Kampus Kiel, Kiel, Germany, 4Klinik für Hämatologie, Onkologie, Immunologie, Palliativmedizin, Infektiologie und Tropenmedizin, München, Germany 1

Introduction: The combination of Bendamustine plus Rituximab (BR) is indicated in treatment-naïve as well as relapsed/refractory (R/R) patients with CLL. The immunmodulatory drug lenalidomide (L) has proofed efficacy in heavily pre-treated CLL. This phase I/II trial was initiated to assess safety/efficacy of the combination BRL in previously untreated (1st-line) and R/R patients with CLL. Patients and methods: Seventeen R/R and five previously untreated medically fit patients with CLL (CIRS score < = 6) were enrolled to receive the combination of BRL. In the R/R phase I- cohort, four different dose levels of L (maximum 15 mg per day [pd]) were tested. In the 1st-line cohort, L was dose escalated from 5 to 15 mg pd. B was administered intravenously (i.v.) with a dose of 50/90 mg/m2 (R/R/1st-line) on day 1 & 2 of each 28 day cycle while R i.v. was dosed with 375 mg/m(2) on day 0 of the first cycle and 500 mg/m(2) on day 1 during subsequent cycles for up to six cycles. Results: The maximal tolerable dose of L determined in the phase I-part of the R/R cohort was 5 mg pd. Overall response rate was 47.1% in the R/R and 60% in the 1st-line cohort, respectively. Median progression-free survival (PFS) was 8.0 months for both cohorts. Median overall survival was 22.9/12.3 months (R/R/1st-line). Grade 3/4 hematologic toxicity was observed in 75% of the R/R- and in 60% of patients receiving 1st-line treatment. Severe infections CTC grade 3–4 occurred in 60% and 43.8% of the patients respectively. Due to high toxicity and low remission rate of BRL the trial was closed prematurely. Conclusion: BRL was associated with a high rate of severe infections, a low response rate and short PFS compared to other 1st- and 2nd-line regimens for CLL and cannot be considered an appropriate treatment option for those patients. Disclosure: No conflict of interest disclosed. V293

Outcomes of anticoagulant (AC) or antiplatelet (AP) use in patients (pts) with chronic lymphocytic leukemia (CLL) or indolent non-Hodgkin’s lymphoma (iNHL) in idelalisib (IDELA) trials Stilgenbauer S.1, Barrientos J.2, Ghia P.3, Pagel J.4, Salles G.A.5, Sharman J.P.6, Gurtovaya O.7, Kim Y.7, Philip B.7, Zelenetz A.D.8 Universität, Ulm, Germany, 2Hofstra North Shore-LIJ School of Medicine, Hempstead, United States, 3Università Vita-Salute San Raffaele and Instituto Scientifico San Raffaele, Milano, Italy, 4University of Washington Fred Hutchinson Cancer Research Center Medical Oncology, Seattle, United States, 5 Université Claude Bernard, Pierre Bénite, France, 6Willamette Valley Cancer Institute and Research Center, Springfield, United States, 7Gilead Sciences, Inc., Foster City, United States, 8Memorial Sloan Kettering Cancer Center, New York, United States 1

Introduction: IDELA, a selective oral PI3Kδ inhibitor, is approved for use in relapsed CLL (in combination with rituximab [R]) and iNHL (as monotherapy). Both diseases occur mainly in the elderly, who have comorbidities that increase thrombotic risk. This post hoc analysis characterized

Abstracts


CONTENTS AUTHOR INDEX

AUTHOR INDEX

the use and outcomes of AC/AP therapy, which was allowed in IDELA registrational clinical trials. Methods: In the phase 3 Study 312–116 (NCT01539512), frail pts with relapsed CLL (including those with any degree of thrombocytopenia) were randomized to receive a combination of continuous IDELA 150 mg BID or placebo (PBO) with 8 R doses. In the phase 2 Study 101–09 (NCT01282424), pts with refractory iNHL received IDELA 150 mg BID until disease progression or unacceptable toxicity. Grade 1, 2, and ≥3 bleeding events were analyzed using MedDRA preferred terms and CTCAE. Results: The 2 trials included 343 pts. In the CLL study, 18 pts (16%) on IDELA + R and 31 (29%) on PBO + R had grade ≥3 thrombocytopenia at baseline. Concomitant AC/AP use was frequent (45% in each study); the most common were aspirin, enoxaparin, and warfarin. AC/AP use was more frequent in pts treated with IDELA + R vs PBO + R. The incidence of bleeding events was similar with IDELA, IDELA + R, and PBO + R. Grade ≥3 bleeding events occurred in 1 IDELA + R, 1 PBO + R, and 3 IDELA pts. Conclusions: AC/AP use involved 45% of the IDELA registrational trial population. Overall, rates of bleeding events were moderate and similar with IDELA + R vs IDELA + PBO; grade ≥3 events were uncommon. There was no specific trend with regard to AC/AP and bleeding events in the 2 arms of the CLL study. Tab. 1. AC or AP use in IDELA clinical trials

n (%)

CLL IDELA+R 110

CLL PBO+R 108

iNHL IDELA 125

Pts on AC/AP

60 (55)

38 (35)

56 (45)

Pts with ≥1 bleeding event (any grade)

15 (14)

20 (19)

17 (14)

Grade 1/2

14 (13)

19 (18)

14 (11)

Event at any time

10 (17)

6 (16)

14 (25)

Event on AC/AP

7 (12)

5 (13)

8 (14)

AC at any time

9

4

13

AP at any time

5

3

6

Pts not on AC/ AP

50

70

69

Event at any time

5 (10)

14 (20)

3 (4)

Disclosure: Stephan Stilgenbauer: Advisory Role: Gilead Sciences; Financing of Scientific Research: Gilead Sciences; Expert Testimony: Gilead Sciences. Andrew Zelenetz: Advisory Role: Gilead Sciences; Expert Testimony: Gilead Sciences. V294

Ibrutinib as salvage therapy for relapsed high-risk chronic lymphocytic leukemia (CLL) in patients after allogeneic hematopoietic stem cell transplantation (HSCT) Hahn M.1, Dietrich S.1, Görner M.2, Welslau M.3, Zenz T.1, Rummel M.4, Schmitt M.1, Köhler A.5, Ho A.D.1, Dreger P.1 Universität Heidelberg, Medizinische Klinik V, Heidelberg, Germany, 2Klinikum Bielefeld, Klinik für Hämatologie, Onkologie und Palliativmedizin, Bielefeld, Germany, 3Klinikum Aschaffenburg, Praxis für Onkologie, Aschaffenburg, Germany, 4Universitätsklinikum Gießen und Marburg, Medizinische Klinik IV, Hämatologie, Gießen, Germany, 5Asklepiosklinik Langen, Praxis für Hämatologie und Onkologie, Langen, Germany 1

Introduction: Patients with high-risk CLL relapsed after HSCT have an overall poor prognosis. The upcoming era of small molecule inhibitors may offer new therapeutic options in that situation.

Abstracts

Study design and patients: In a single centre retrospective analysis, the outcome of ibrutinib salvage therapy was assessed in patients with highrisk CLL relapsed after HSCT. Results: Ibrutinib was administered to 8 patients (m/f= 1/1) with a median age of 53y at ibrutinib start. 7/8 had been transplanted from an unrelated donor (6 MUD, 1 MMUD), 4/8 were 17p- at relapse. The median time from HSCT to ibrutinib start was 33 (5–88) months. Ibrutinib was the primary relapse therapy in 4 patients. The remainder received ibrutinib after failure of pretreatment with rituximab + DLI (2 patients) or with rituximab + DLI followed by rituximab + lenalidomide (2 patients). With a median therapy duration of 7 (1–14) months, no grade 3/4 toxicity has been observed. All 8 patients are in ongoing response: Six patients have accomplished a partial remission (PR), one patient being under therapy only since one month reported symptom relief, and one patient has achieved an MRD-negative complete remission (CR). This patient had primary DLI failure and subsequently received ibrutinib for lowering of tumor burden, while DLI applications were continued. After having achieved CR upon simultaneous treatment with ibrutinib and DLI, the patient developed cGvHD and for the first time became MRD-negative. Another patient had been transplanted in an ibrutinib-induced CR. After ibrutinib had been discontinued in the context of HSCT, an early relapse with rapidly rising leucocyte count ocurred 72d post HSCT. As a consequence, immunosuppression was tapered and ibrutinib treatment was resumed immediately after immunosuppression withdrawal, which resulted in a PR with normalized leucocyte count after 4 months. Conclusion: This preliminary data suggest that ibrutinib is a safe and effective therapy for relapse of high-risk CLL after HSCT, even in patients pre-exposed before HSCT. Ibrutinib seems to allow for GvL activity and might serve as a priming therapy for DLI. Further studies are needed to decipher the immunologic interactions between ibrutinib and the allograft, and to analyze long-term outcome of patients treated with ibrutinib for relapsed high risk CLL. Disclosure: No conflict of interest disclosed. V295

CLL – patient registries confirm bendamustine-containing regimen (BR) as an effective first-line therapy Linde H.1, Göttel R.2, Tessen H.-W.3,4 Medizinisches Versorgungszentrum für Blut- und Krebserkrankungen, Potsdam, Germany, 2rgb Onkologisches Management GmbH, Sarstedt, Germany, 3Onkologische Kooperation Harz, Goslar, Germany, 4Projektgruppe Internistische Onkologie (PIO), Goslar, Germany 1

Introduction: The chronic lymphocytic leukaemia (CLL) is the most common leukemic disease in Central Europe. The median age of onset is between 70 and 75 years. The combination of bendamustine and rituximab has proven effective for the treatment of this disease in clinical trials and everyday use. Methods: Since 2008, 61 hemato-oncological practices from 16 federal states within the project team of internal oncology (PIO) have been documenting disease histories of patients with chronic lymphocytic leukaemia in the registry ONCOReg. 769 patients received a bendamustine-containing therapy, 525 (68.3%) as first-line therapy, 124 (23.6%) of which bendamustine mono and 401 (76.4%) bendamustine/rituximab (BR). Results: This analysis presents the results of the use of bendamustine/ rituximab in the first-line treatment of CLL patients in clinical practice. Patient characteristics: Gender: 66% m / 34% f Median age: 71 years Without B-symptoms: 61% ECOG 0/1/2: 26% / 59% / 15% BINET A/B/C: 11% / 55% / 34% Period from initial diagnosis until first therapy: 22.3 months Comorbidities: 37% hypertension, 19% diabetes; 10% CHD, 8% AIHA/ITP

Oncol Res Treat 2015;38(suppl 5):1–270

93


CONTENTS AUTHOR INDEX

AUTHOR INDEX

Therapy: A median of 6 (1–8) cycles were administered. The median total dose of bendamustine was 880 mg/m². Response: The objective remission rate is at 88.8%, 48% of which CR (haematologically tested) and 41% PR. Survival: The median follow up is 24.3 months. The median progression-free survival is at 44.5 months. The median overall survival has not been reached yet. The 3-year survival rate is at 84%. Conclusions: The analysis of the registry reflects the treatment routine and reveals the treatment of mostly older comorbid patients. The therapy with bendamustine in combination with rituximab proves to be highly effective and safe. The remission rates and PFS of ONCOReg are comparable to those of other patients registries (TLN) or to clinical studies such as CLL2M or CLL10. Recent studies in which older and comorbid (“not fit”) patients were treated with newer substances do not show better results with regard to CR and PFS than the combination BR. The results emphasize the high quality of the treatment of patients in the everyday routine and in specialized oncology practices. Disclosure: Hartmut Linde: Expert Testimony: Honorare für Teilnahme an Studien (Firma Mundipharma, Roche, Universität Köln); Other Financial Relationships: Übernahme von Fortbildungs-, Reise- und Beherbergungskosten (Firma Mundipharma, Roche) Hans-Werner Tessen: No conflict of interest disclosed.

Fortbildung

Ethik Handeln am Lebensende V296

Physicians´ practice at the end of life. Empirical data, ethical challenges Schildmann J.1 Ruhr-Universität Bochum, Institut für Medizinische Ethik und Geschichte der Medizin, Bochum, Germany 1

Introduction: Ethical questions at the end of life are at the centre of scientific and public debates. Up to the present there is scarcity of empirical data regarding physicians’ end-of-life practices in Germany. Methods: Cross-sectional study among a random sample of German physicians by means of the questionnaire of the EURELD Consortium with additional questions on experiences and attitudes regarding physician assisted suicide. Results: 734 physicians from five state chambers of physicians responded (response rate 36.9%). 403 physicians reported about end-of-life practices regarding adult patients. Alleviation of symptoms took place in 86.7% of cases and in 50.7% medical treatment had been withheld. 30.8% of patients received continuous sedation till death. In three cases death was the consequence of a drug which was provided or administered by respondents. 20.7% of respondents had been requested to perform physician assisted suicide (PAS). 41.7% of participants could not imagine participating in PAS, whereas 40.2% could imagine this under certain circumstances. A prohibition of PAS by professional law war rejected by 33.7%, 25.0% support such a ban and 41.4% were undecided. Conclusion: The published data will be presented together with data of an online survey among members of the German Society for Hematology/Oncology on their experiences and views regarding physician assisted suicide. The relevance of such data for the current societal and political debate and as starting point for reflections on ethical issues at the end of life in cancer care will be discussed. Disclosure: No conflict of interest disclosed.

V298

The role of physicians in the area of assisted suicide. Legal and medical ethical standards in Switzerland Güth U.1 Kantonsspital Winterthur, Department Geburtshilfe & Gynäkologie, Brustzentrum Senosuisse, Winterthur, Switzerland 1

Unlike in most European countries, physician-assisted suicide is not illegal in Switzerland, insofar as the actions of the person are not in self-interest (Art. 115 of the Swiss Penal Code). The number of assisted suicides aided by right-to-die organizations such as Exit or Dignitas has sharply increased in the last 20 years. Approximately half of the cases of assisted suicides are comprised of people with late stage cancer; the second most common reason for choosing assisted suicide is affliction with neuro-degenerative diseases (above all multiple sclerosis, amyotrophic lateral sclerosis and Parkinson’s disease). According to current data, approximately 25% of the officially reported suicides in Switzerland are physician-assisted. Physician-assisted suicide is widely accepted in the Swiss society. In the canton of Zurich in 2011, a petition to ban assisted suicide was rejected by 85% of voters in a referendum. The Schweizerische Akademie der Medizinischen Wissenschaft (SAMW) rejects assisted suicide as a physician`s duty, because it contradicts the basic principals of medicine. Nevertheless, it concedes that a doctor’s conscientious and personal decision to perform assisted suicide for individual terminally-ill patients should be respected. The central part of the doctor’s involvement in assisted suicide is the prescription of a lethal dose of sodium pentobarbital. In doing so, the doctor has to apply to the rules of due medical care. A prescription can only be issued if the patient has been examined by the physician concerned. The patient must be informed about his diagnosis, about the expected prognosis, and about different alternative treatment options, in particular about the possibilities offered by palliative care. According to the SAMW, assisted suicide is only possible when the end of life is in the foreseeable future and the person is capable of making the decision willingly on his own without outside influences or pressure. A third party with the capability to assess whether these criteria have been met must confirm the decision before assisted suicide can legally be performed. The practice of assisted suicide in Switzerland is characterized by the close cooperation between medical and non-medical personnel. In general, a staff member of the right-to-die organization, but not the doctor, is present during the suicide. Following death, the assisted suicide has to be reported to the authorities as a death under special circumstances. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Lymphome aggressiv experimentell V299

USP9X promotes resistance to spindle poisons in aggressive B-cell lymphoma by regulating the mitotic cell fate decision Engel K.1, Rudelius M.2, Altmann B.3, Abhari B.A.4, Brunner A.1, Kurutz J.1, Targosz B.-S.1, Loewecke F.1, Knorn A.-M.1, Fernandez-Sáiz V.1, Baumann U.1, Glöckner J.5, Pfreundschuh M.6, Trümper L.7, Jost P.1, Klapper W.8, Fulda S.4, Peschel C.1, Bassermann F.1 III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität, München, Germany, 2Pathologisches Institut/Universität, Würzburg, Germany, 3 IMISE, Leipzig, Germany, 4Institut für Experimentelle Tumorforschung in der Pädiatrie, Frankfurt, Germany, 5DZNE, Tübingen, Germany, 6Klinik für Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany, 7Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany, 8Institut für Pathologie, Kiel, Germany 1

Introduction: Despite advances in the first line therapy, a significant number of patients with aggressive B-cell lymphoma succumb to relapsing or refractory disease. The mitotic spindle assembly checkpoint (SAC)

94

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

is an important target of lymphoma therapy as it regulates resistance to spindle poisons. To date it has remained unclear how cells execute cell fate decisions under conditions of SAC induced mitotic arrest. Methods: Binding and stability assays were performed using immune precipitation, induced expression or siRNA-mediated knockdown. Ubiquitylation was assessed under denaturing conditions. Analysis of cells synchronized with thymidine and nocodazol or paclitaxel was supported by two dimensional cell cycle analysis using FACS (BrdU, PI). BL57 mice were injected with lentivirally transduced Eµ-myc lymphoma cells. Patient samples were obtained from the German High Grade Non Hodgkin’s Lymphoma Study Group (DSHNHL, Ricover-60 Trial) and analysed by immunohistochemistry. Results: We identify USP9X as the mitotic deubiquitinase of the X-linked inhibitor of apoptosis protein (XIAP), and demonstrate that deubiquitylation and stabilization of XIAP by USP9X induces resistance towards mitotic spindle poisons[FB1] . Mitotic cell death in response to prolonged taxol exposure is increased by USP9X knockdown in WT, cIAP2 k.o. and MCL-1 k.o. but not in XIAP k.o. MEFs, indicating that XIAP marks the biologically relevant target of USP9X. Mapping experiments delineate the USP9X binding site in XIAP to the Gly188 residue. Accordingly, overexpression of USP9X and XIAP, but not of instable XIAP(G188) mutants, promote cell survival despite taxol exposure. USP9X-XIAP overexpressing DLBCL lines exhibit increased resistance to spindle poisons and undergo substantial apoptosis in response to USP9X knockdown or pharmacological inhibition and treatment with the XIAP inhibiting SMAC mimetic BV6. Likewise, knockdown of USP9X or XIAP delays lymphoma development in a murine Eµ-myc-lymphoma model. In 121 human DLBCL samples high expression of USP9X significantly correlated with XIAP overexpression (p = 0.004) and overexpression of USP9X and XIAP was associated with significantly reduced event free survival in CHOP treated DLBCL patients (p = 0.05). Conclusion: Antiapoptotic signaling via the USP9X-XIAP axis mediates resistance to mitotic cell death. USP9X-XIAP expression may serve as an attractive novel prognostic and therapeutic target in aggressive B-cell lymphoma. Disclosure: Katharina Engel: No conflict of interest disclosed. Florian Bassermann: Expert Testimony: Celgene. V300

Vitamin D promotes tumoricidal activity of macrophages and improves the efficacy of antibody-dependent cellular cytotoxicity Bruns H.1, Büttner M.2, Mougiakakos D.2, Bittenbring J.3, Beier F.4, Pasemann S.2, Fabri M.5, Neumann F.3, Mackensen A.2, Gerbitz A.2 Uniklinikum Erlangen, Dept. of Internal Medicine 5 – Hematology/Oncology, University of Erlangen, Erlangen, Germany, 2Uniklinikum, Erlangen, Germany, 3 Universitätsklinikum des Saarlandes, Homburg, Germany, 4Uniklinik, Aachen, Germany, 5Uniklinik, Köln, Germany 1

Infiltration by macrophages represents a characteristic morphological hallmark in high-grade lymphatic malignancies such as Burkitt’s lymphoma (BL). Although macrophages can, in principle, target neoplastic cells and mediate antibody-dependent cytotoxicity (ADCC), tumor-associated macrophages (TAMs) regularly fail to exert direct cytotoxic functions. The underlying mechanisms responsible for this observation remain unclear. Here, we demonstrate that inflammatory M1 macrophages kill proliferating high-grade B cell lymphoma cells by releasing the antimicrobial peptide cathelicidin in a vitamin D-dependent fashion. We show that cathelicidin directly induces cell death by targeting mitochondria of BL cells. In contrast, anti-inflammatory M2 macrophages and M2-like TAMs in BL exhibit an altered vitamin D metabolism, resulting in a reduced production of cathelicidin and consequently in inability to lyse BL cells. However, treatment of M2 macrophages with the bioactive form of vitamin D, 1,25D3, or a vitamin D receptor agonist effectively induces cathelicidin production and triggers tumoricidal activity against BL cells. Furthermore, rituximab-mediated cytotoxicity of vitamin D-treated M2

Abstracts

AUTHOR INDEX

macrophages is cathelicidin-dependent. Finally, vitamin D treatment of 25D-deficient volunteers in vivo or primary TAMs in vitro improves rituximab-mediated ADCC against B cell lymphoma cells. These data indicate that activation of the vitamin D signaling pathway activates antitumor activity of TAMs and improves the efficacy of ADCC. Disclosure: No conflict of interest disclosed. V301

Arylindolylmalemide PDA-66 and Its derivate PDA-377 show antiproliferative effects on B-cell non-Hodgkin lymphoma cell lines with MYC and BCL rearrangements van der Wall K.1, Roolf C.1, Sklarz L.-M.1, Eschenburg A.1, Sekora A.1, Murua Escobar H.1, Rolfs A.2,3, Pews-Davtyan A.4, Beller M.4, Junghanss C.1 Universitätsmedizin Rostock, Klinik III, Hämatologie, Onkologie, Palliativmedizin, Rostock, Germany, 2Universitätsmedizin Rostock, AlbrechtKossel-Institut für Neuroregeneration, Rostock, Germany, 3Centogene AG, Rostock, Germany, 4Universität Rostock, Leibniz-Institut für Katalyse, Rostock, Germany 1

Introduction: Diffuse large B-cell lymphoma (DLBCL) as well as Burkitt`s lymphoma are a frequent subtypes of non-Hodgkin´s lymphoma (NHL). Both subtypes usually are characterized by short cell doubling times making them susceptible to cell cycle modifying agents. Previous own results showed that the novel arylindolmaleimide PDA-66 inhibits the polymerisation of microtubules inducing antiproliferative effects in acute lymphoblastic leukemia cells. Here, we evaluated the potency of PDA-66 and its derivate PDA-377 on B-cell NHL cell lines. Methods: Burkitt lymphoma and DLBCL cell lines with BCL2 and MYC rearrangements (DOGKIT; SU-DHL-4 and WILL-2) were incubated for 72 h with increasing concentrations ranging from 1 to 5 µM of PDA-66 and PDA-377, respectively. Metabolic activity was determined by WST-1 assay. Furthermore, PDA-66 was combined with low dose of AraC, Doxorubicin (Doxo) or Dexamethasone (Dexa) in different sequences (PDA before, concomitant, after 2nd drug). Changes in cell cycle and apoptosis were analyzed flow cytometricaly by propidium iodide and annexin staining. Results: In all cell lines PDA-66 and PDA-377 inhibited proliferation significantly in a dose dependent manner. Strongest antiproliferative effects were detected with PDA-66. Incubation with 5 µM PDA-66 decreased metabolic activity to 22.4% in WILL-2, 47.1% in SU-DHL-4 and 43.3% in DOGKIT cells. Further, treatment with PDA-66 resulted in an increase of cells arresting in G2/M phase of the cell cycle (41.4% vs. control: 14.1%). This effect was enhanced when PDA-66 and Dexa were combined together (68.9% in G2/M) even when cells were less sensitive to single Dexa. Interestingly, the sequence of PDA-66 and Dexa application to DBCL cells had an impact on the antiproliferative effects. A 24 h pre-treatment with PDA-66 (2.5 µM) before adding Dexa (50 µM) increased antiproliferative effects compared to other sequences. Conclusion: Treatment with PDA-66 and PDA-377 induced antiproliferative effects on B-NHL harbouring BCL2 or MYC gene rearrangements. Moreover, PDA-66 seems to sensitize DLBCL cells to Dexa. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

95


CONTENTS AUTHOR INDEX

AUTHOR INDEX

V302

V303

Identification of a N-hyperglycosylated domain of CNS proteins as B cell receptor antigen in primary CNS lymphoma

Functional investigation of putative genetic lesions on the CNS tropism of diffuse large B-cell lymphomas in vivo

Thurner L.1, Kemele M.1, Fadle N.1, Regitz E.1, Roth P.2, Weller M.2, Schäfer H.3, Schorb E.3, Illerhaus G.3, Szczepanowski M.4, Klapper W.4, Monoranu C.-M.5, Rosenwald A.5, Buslei R.6, Bohle R.M.7, Preuss K.-D.1, Pfreundschuh M.1

Reimann M.1, Maßwig S.1, Schleich K.1, Herrmann A.1, Lohneis P.2, Schrezenmeier J.F.1, Dörken B.1,3, Schmitt C.1,3

Saarland University Medical School, José Carreras Center for Immuno- and Gene Therapy and Internal Medicine, Homburg/Saar, Germany, 2Department of Neurology, University Hospital, Zürich, Switzerland, 3Department of Hematology and Oncology, University Medical Center, Freiburg, Germany, 4 Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany, 5Institute of Pathology, University of Würzburg, Würzburg, Germany, 6Institute of Neuropathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany, 7Saarland University Medical School, Institute of Pathology and Neuropathology, Homburg/Saar, Germany 1

Background: A reported VH4–34 bias in PCNSL has raised speculations of a possible chronic stimulation of BCR by a CNS-autoantigen. The present study focused on the search for these hypothetic autoantigens in immunocompetent patients. Methods: Recombinant Fabs were expressed based on corresponding pairs of functional variable region heavy and light chain genes, which had been amplified from snap-frozen lymphoma specimens by semi-nested PCRs. Subsequently these rec. Fabs were screened on protein arrays. Results: Rec. Fab expression was attempted in 21 and successful in twelve PCNSL cases. Protein array screening of the rec. Fabs revealed that 8 of 12 rec. PCNSL-BCRs reacted with SAMD14 and the SAM domain of neurabin-1, two proteins with high homology and preferential expression in the CNS. Proteomic analysis of lymphoma specimens showed that SAMD14 and the SAM domain of neurabin-1 were alternatively N-glycosylated in patients with a Fab-specificity against SAMD14/neurabin-1, but not in the remaining cases with other BCR specificities. Atypical (N-L-E-Q) instead of (N-X-S/T) N-hyperglycosylations of SAMD14/neurabin-1 were shown for every case with sufficient biopsy material for this proteomic analysis and a PCNSL-BCR specific for SAMD14/ neurabin-1. Of the rec. BCRs of all cases with sufficient material to test for hyperglycosylation, only the rec. Fabs derived of the cases with hyperglycosylated SAMD14/neurabin-1 reacted against SAMD14/neurabin-1. No N-hyperglycosylations of SAMD14/ neurabin-1 were neither detected in the peripheral blood of 400 healthy controls, 100 newborns, 50 nursery home residents nor in 86 established cell lines of various cellular origin. Moreover, antibodies against SAMD14/neurabin-1 were detected in the sera and cerebrospinal fluids of an independent second cohort of patients with PCNSL (8/22), but not in sera of patients with secondary CNS manifestations of systemic DLBCL (0/17) or of healthy controls (0/92). Conclusion: Our results suggest that atypical (NLEQ) glycosylation of the highly homologous SAM domain of SAMD14 and neurabin-1 maintains a chronic autoantigenic stimulation in the CNS, ultimately leading to the malignant transformation of B-cells with a BCR specific for these atypically N-hyperglycosylated proteins into an aggressive B-cell lymphoma in the CNS, providing strong evidence for the role of chronic autoantigenic stimulation as a first step in the pathogenesis of aggressive B-cell lymphomas. Disclosure: No conflict of interest disclosed.

Charité – Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumor Immunology and Molekulares Krebsforschungszentrum, Berlin, Germany, 2Charité – Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany, 3Max-Delbrück-Center for Molecular Medicine, Berlin, Germany 1

Introduction: Secondary central nervous system (CNS) lymphomas impose a dismal prognosis in patients suffering from systemic diffuse large B-cell lymphomas (DLBCL). Myc rearrangement, chromosomal deletions at the CDKN2A (a.k.a. INK4a/ARF) or ATM gene loci as well as recurrent NF-kB-hyperactivating mutations are frequent in CNS-tropic lymphoma, however, investigations that functionally validate these lesions in adequate in vivo model systems are missing. Methods: We generated primary Eµ-myc transgenic lymphomas with distinct naturally occurring NF-kB mutations (within genes encoding for MyD88, CD79B, A20, IkBζ, IkBε or BIRC3) or deletions at the INK4a/ ARF and ATM loci. Wild-type recipient mice were i.v. transplanted with these lymphoma cells, and subsequently monitored for systemic lymphoma development, at which the brain was isolated and examined regarding lymphoma infiltration. Results: Underscoring Myc’s role as a putative co-driver of CNS involvement, we found in about 40% of primary Eµ-myc lymphomas (with no additional exogenous lesions) meningeal CNS tropism. Genome-wide transcriptome analysis unveiled NF-kB hyperactivation in the CNS-tropic lymphoma group, suggesting that NF-kB-activating mutations promote CNS-prone pathogenesis in vivo. Transplantation of Eµ-myc hematopoietic stem cells expressing a variety of NF-kB-activating mutants resulted in a significant acceleration of Eµ-myc-driven lymphomagenesis, with some, but not all of these mutants conferring a CNS-tropic lymphoma phenotype. Global NF-kB suprression in CNS-tropic Eµ-myc lymphomas via the NF-kB-antagonizing IkBΔN super-repressor did not fully abrogate lymphoma infiltration of the brain, suggesting that additional factor(s) must contribute. Importantly, targeted ablation of the INK4a/ARF and ATM loci robustly enhanced CNS tropism of Eµ-myc lymphomas. Conclusions: The Eµ-myc mouse lymphoma model is well-suited to genetically dissect and rebuild components of CNS tropism. We identified CDKN2A or ATM deletions as critical determinants of CNS tropism in vivo. Our systematic analyses of different NF-kB mutants – so far rather recognized as functionally interchangeable – indicated that only distinct NF-kB mutants contribute to CNS tropism in B-cell lymphomas. Our findings underscore the need for functional analyses of oncogenic network contexts, and provide important insights into candidate target lesions for personalized CNS-directed therapies in DLBCL patients in the future. Disclosure: No conflict of interest disclosed. V304

A modified autoantigen is the first molecularly defined risk factor and a dominant antigenic target / stimulus of the B-cell receptor from ABC-type DLBCL Pfreundschuh M.1, Preuss K.-D.1, Fadle N.1, Regitz E.1, Kemele M.1, Bohle R.-M.2, Hansmann M.-L.3, Thurner L.1 Universität des Saarlandes, Innere Medizin I, Homburg/Saar, Germany, Universität des Saarlandes, Institut für Allgemeine und Spezielle Pathologie, Homburg/Saar, Germany, 3Universitätsklinikum Frankfurt, Dr. Senckenbergisches Institut für Pathologie, Frankfurt am Main, Germany 1 2

Introduction: Chronic antigenic stimulation may play an important role in the pathogenesis of malignant lymphomas. We have previously shown that hyperglycosylated neurabin/SAMD14 is the antigenic target of the B-cell receptor (BCR) of 2/3 of all primary CNS lymphomas, but BCR for peripheral DLBCL have not been defined to date.

96

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Methods: BCRs were expressed as recombinant Fabs based on corresponding pairs of functional variable region heavy and light chain genes, which had been amplified from isolated genomic DNA of snap-frozen lymphoma specimens and DLBCL-derived cell lines. The purified BCRFabs were checked for binding to proteins expressed on macroarrays of human cDNA expression libraries. Results: The BCR from 10 DLBCL cell lines (5 of the germinal center type and 5 of the activated B-cell type) were tested on the protein macroarray. None of the GC-type BCR reacted with any of the proteins expressed on the protein macroarray, but the BCR from 3/5 (60%) of the ABC-derived cell lines reacted with ARS2 (arsenite resistance protein 2), a conserved mammalian protein which is important for microRNA biogenesis. Isoelectric focusing and phosphatase treatment of ARS2 derived from ABC cell lines with a BCR specific for ARS2 revealed that ARS2 was hypophosphorylated (hypo-ARS2) in the respective cell lines. Analysis of peripheral blood lymphocytes from patients with DLBCL of unknown cell of origin and healthy controls revealed that 5/100 (5%) of patients, but only 1/400 (0.25%) of controls were carriers of hypo-ARS2, resulting in a 20x increased risk for healthy carriers of hypo-ARS2 to develop DLBCL. All patients with BCRs targeting ARS2 had polyclonal antibodies against ARS2 in their serum. Conclusions: Hypo-ARS2 is the first molecularly defined risk factor for DLBCL identified to date. The increased risk for healthy carriers of this posttranslational modification to develop DLBCL supports the hypothesis of chronic antigenic stimulation as an important factor in the pathogenesis of DLBCL and indicates that posttranslationally modified autoantigens are a frequent target and stimulus for DLBCL-BCR. That antibodies against ARS2 are found in the respective patients suggests that the DLBCL evolves from a polyclonal B-cell response against this autoantigen. Investigations into the mechanism underlying the hypophosphorylation of ARS2 are underway and therapeutic consequences will be discussed. Supported by Wilhelm-Sander-Stiftung. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Tumor-/Zellbiologie I V305

The tumor-supporting role of oxidative and ER stress tolerance: New lessons from fibroblasts and cancer cells to understand tumor biology and chemoresistance Venkataramani V.1, Rumkamp T.1, Pape V.1, Kiecke C.1, Küffer S.2, Ströbel P.2, Trümper L.1, Wulf G.G.1 University Medicine Goettingen, Department of Hematology and Oncology, Göttingen, Germany, 2University Medicine Goettingen, Department of Pathology, Göttingen, Germany 1

Introduction: Conversely to untransformed cells, where deregulated cellular stress conditions drive the activation of death pathways, malignant cells exploit oxidative milieu for its advantage. Therefore, resistance to oxidative stress appears to be a major mechanism of cancer initiation/ propagation and chemoresistance. In this regard, we have identified a novel role for the redox modulator amyloid precursor protein (APP) in effectively modulating the stress tolerance of cancer cells. We previously established that APP is overexpressed in several solid and haematological cancer types and underlined the function in the cell proliferation, colony formation and propagation of cancer stem cells. Now, we show that downregulation of APP in cancer cells results in a feed-forward loop with oxidative stress and its induction of endoplasmic reticulum stress (ER stress). The resulting selective DNA damage response (DDR) impacts growth of cancer cells/transformed fibroblasts and confers sensitization especially to genotoxic chemotherapeutics agents. Methods: Murine embryonal fibroblasts (MEFs) from wild-type (APP+/+) and APP–/– (APP-knockout) mice were isolated and trans-

98

Oncol Res Treat 2015;38(suppl 5):1–270

formed with Ha-RasV12 via adenoviral transfer. Functional assays such as cell proliferation, colony forming assays, RNA seq, and western blot as well as in ovo analysis via CAM-Assay were performed. Intracellular ROS and lipidperoxidation levels were determined via DCFH-DA dye and BODIPY 581⁄591 C11 dye, respectively. Results: Consistent with our hypothesis that APP modulates intracellular iron homeostasis, we show that loss-of-APP either in cancer cells or APP-/- MEFs contain higher ROS and lipidperoxidation levels. Beside oxidative stress, we provide direct genetic evidence that APP causes cell death in a pathologically relevant form leading to ER stress inducing DNA damage. Novel selective APP-Inhibitors also induced ROS and ER-stress causing activation of DDR without inducing apoptosis or autophagy. By dissecting the molecular hallmark events, we present evidence that lossof-APP suppresses tumorigenicity of transformed fibroblasts and reveal that APP-induced alteration of the DNA damage-signaling effectively re-sensitize several cancer types to various cancer therapies, including DNA damaging agents. Conclusion: Here, we identify APP as regulator of oxidative and ER stress selectively modulating DNA damage signaling representing an effective drug target for enhanced cancer therapy. Disclosure: No conflict of interest disclosed. V306

Inhibition of PARP selectively sensitizes KRAS-mutant cancer cells to chemotherapy in vivo – an update Hähnel P.S.1, Sasca D.1, Enders B.1, Lehmann N.1, Roos W.P.2, Kaina B.2, Theobald M.1, Kindler T.1 University Medical Center of the Johannes Gutenberg-University, Third Department of Medicine, Division of Hematology, Medical Oncology & Pneumology, Mainz, Germany, 2University Medical Center of the Johannes Gutenberg-University, Institute of Toxicology, Mainz, Germany 1

Introduction: Activating KRAS mutations are detected in a substantial number of different malignancies and often appear during early stages of tumorigenesis. KRAS-mutations are already found in patients suffering from colorectal adenomas or pancreatic ductal hyperplasias. Full malignant transformation is thought to be triggered by additional mutations. In preliminary work we demonstrated that KRAS-mutated cells rely on the alternative non homologeous end-joining (alt-NHEJ) repair pathway upon genotoxic stress, while KRAS wild-type cells do not. RNAi-mediated down regulation of the alt-NHEJ component DNA ligase 3α abolished drug-resistance to apoptosis in KRAS-mutant cells in vitro. In vivo, treatment with the PARP-inhibitor Olaparib sensitized specifically KRAS-mutated cells to chemotherapeutic agents. Combination therapy with Irinotecan or VP-16 was able to control tumor growth in NSG mice transplanted with colon cancer and NSCLC cells, respectively. Methods: The sensitivity to drug-induced apoptosis of different colon cancer and NSCLC cell lines with either mutated or wild-type KRAS was analyzed by molecular assays and FACS analysis. Cell viability and cytotoxicity were determined via cell proliferation assays and cell cycle analysis. In vivo, the antitumor activity of Olaparib in combination with different genotoxic agents was assessed in mice bearing tumor xenografts. Results: Pharmacologic inhibition of PARP, which has been implicated in promoting end joining by alt-NHEJ, upon treatment with Olaparib caused increased apoptosis induced by standard genotoxic agents compared to controls. Mice bearing tumor xenografts either from colon cancer or NSCLC cell lines showed a diminished tumor growth compared to animals treated with monotherapy, finally resulting in longer overall survival. Interestingly, the reversal of genotoxic drug resistance upon inhibition of the alt-NHEJ pathway seems to be specific for KRAS-mutant cells, as no effect was observed in tumors derived from KRAS-wild-type cell lines. Conclusion: Our data provide evidence for a synthetic lethal interaction between KRAS-mutations and DNA damage repair. Furthermore, they indicate that targeting components of the alt-NHEJ pathway, e.g., PARP, sensitizes KRAS-mutant cancer cells to standard chemotherapeutics and

Abstracts


CONTENTS AUTHOR INDEX

represents a promising approach for inducing synthetic lethal vulnerability in cells harboring otherwise non-druggable KRAS mutations. Disclosure: No conflict of interest disclosed. V307

Loss of the tumor- and metastasis suppressor RAF kinase inhibitor protein is caused by the overexpression of miRNA-23a Hatzl S.1, Geiger O.1, Kuepper M.K.1, Seime T.2, Nußbaumer E.1, Wieser R.3, Pichler M.4, Scheideler M.5,6,7, Nowek K.8, Jongen-Lavrencic M.8, Wölfler A.1, Troppmair J.2, Sill H.1, Zebisch A.1 Division of Hematology, Medical University of Graz, Graz, Austria, 2Daniel Swarovski Research Laboratory, Innsbruck Medical University, Innsbruck, Austria, 3Department of Medicine I, Medical University of Vienna, and Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria, 4Division of Oncology, Medical University of Graz, Graz, Austria, 5 Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany, 6German Center for Diabetes Research (DZD), Neuherberg, Germany, 7 University Hospital, Heidelberg, Germany, 8Department of Hematology, Erasmus University Medical Center, Rotterdam, Netherlands 1

Introduction: RAF kinase inhibitor protein (RKIP) is a negative regulator of MAPK/ERK signaling and acts as a tumor- and metastasis suppressor. A complete or partial loss of RKIP expression has been observed in several human malignancies, including acute myeloid leukemia (AML). As the mechanisms leading to RKIP loss are still unclear, we analyzed the potential involvement of miRNAs within this process. Methods: RKIP protein expression was assessed in 33 AML specimens, which had been characterized by miRNA microarrays previously (Rommer et al., BMC Cancer 2013). Chip data of miRNAs related to RKIP expression were further validated by qPCR. U937 cells were transduced with shRNA or overexpression constructs for RKIP using lentiviral transduction. miRNA mimics, as well as an RKIP 3’UTR Luciferase reporter plasmid were transfected by lipofection (in HEK-293) and nucleofection (in NB-4), respectively. qPCR and Western blot were used to monitor the expression of RKIP and miRNAs. Correlation between miRNA-23a and RKIP expression was further corroborated in mRNA and miRNA microarray data from an independent AML cohort (n = 214), as well as in a database retrieval using The Cancer and Genome Atlas (TCGA; AML, n = 173; invasive breast carcinoma, n = 283; renal clear cell carcinoma, n = 171; ovarian serous cystadenocarcinoma, n = 516). Results: By combining RKIP expression with both, miRNA microarray and qPCR data, we identified a set of five miRNAs showing increased expression levels in AML specimens with RKIP loss. Increased expression of miRNA-23a could be corroborated in two independent AML cohorts, comprising almost 400 patients. In functional experiments, modulation of RKIP expression failed to influence miRNA-23a, thereby excluding that miRNA-23a overexpression is an effect of RKIP loss. On the contrary, overexpression of miRNA-23a by miRNA-mimics markedly decreased RKIP mRNA and protein expression, indicating that it is functionally involved in the downregulation of this tumor-suppressor. Preliminary results with an RKIP 3’UTR Luciferase reporter clone suggest, that this effect is mediated by binding of miRNA-23a to this region. Finally, a database retrieval comprising almost 1000 patients with other cancer entities also showing RKIP loss, further corroborated association of miRNA-23a overexpression with decreased RKIP expression, thereby supporting the relevance of this finding. Conclusions: Our data have identified miRNA-23a as a negative regulator of RKIP expression. Disclosure: No conflict of interest disclosed.

Abstracts

V308

Very early functional in-vivo imaging to predict response to B cell receptor pathway targeting in lymphoma Habringer S.1,2, Li Z.2, Pietschmann E.2, Slawska J.2, Walch A.3, Peschel C.2,4, Keller U.1,2 DKFZ/DKTK, Partnerstandort München, München, Germany, 2Technische Universität München, III. Medizinische Klinik, München, Germany, 3Helmholtz Zentrum, München, Germany, 4DKFZ/DKTK, Partnerstandort München, Heidelberg, Germany 1

B cell receptor (BCR) signaling plays a pivotal role for disease development and progression in both activated B cell (ABC) and germinal center B cell (GCB) diffuse large B cell lymphoma (DLBCL). However, there is evidence that ABC DLBCL primarily depends on chronic active BCR signaling with strong NFκB activation, whereas GCB-DLBCL is characterized by tonic BCR signaling, leading to PI3K activation. These differences in BCR signaling impact the response of ABC and GCB DLBCL to kinase inhibitors affecting BCR pathway components. We assessed response to ibrutinib and idelalisib in ABC and GCB cell lines harboring different mutations in the BCR pathway in-vitro and in a xenograft mouse model in-vivo and evaluated the ability of functional imaging technologies to predict the response to these inhibitors. Both idelalisib and ibrutinib inhibited proliferation in the CD79A mutated ABC cell line OCI-Ly10 in-vitro, but had no effect on the TAK1 mutated ABC cell line U2932 and the GCB cell line SUDHL-6, as was predicted by the mutational status of BCR components in these cells. To further analyze if response to BCR signaling inhibitors could be imaged in vivo, we xenotransplanted OCI-Ly10, SUDHL-6 and U2932 cells subcutaneously into SCID mice and administered both inhibitors orally after successful engraftment of lymphoma cells had occurred. To monitor response to therapy, mice were imaged with FDG- and FLT-PET immediately before and 48 hours after initiation of therapy. Both inhibitors induced rapid reduction in tumor volumes of OCI-Ly10 and, surprisingly, U2932 lymphomas, but had no effect on SUDHL-6 tumors, which indicated that mutational status of BCR signaling components alone is not sufficient to predict response to BTK and PI3K inhibition in this in-vivo model. Finally, MALDI imaging was performed on xenograft lymphomas to assess patterns of proteomic changes associated with response or resistance to BCR pathway inhibition. In OCI-Ly10 lymphomas, we found a signature of proteomic changes induced by ibrutinib that separated treated and untreated lymphomas in PCA-clustering. We found that mutational status of BCR components was sufficient to predict response of OCI-Ly10, U2937 and SUDHL-6 to ibrutinib and idelalisib in-vitro, but not in-vivo. MALDI-imaging to detect changes in the proteome could serve as a potential method to predict response or resistance to BCR pathway inhibition early after initiation of therapy. Disclosure: No conflict of interest disclosed. V309

Multi-molecular complexes of Bcl-2 proteins regulate membrane permeabilization by Bax Bogner C.1,2, Kale J.2, Chi X.2, Leber B.2, Andrews D.2 III. Medizinische Klinik Klinikum rechts der Isar, Hämatologie/Onkologie, München, Germany, 2Sunnybrook Research Institute, Biological Sciences, Toronto, Canada 1

The core event in apoptosis is the mitochondrial outer membrane permeabilization (MOMP), the “point of no return” in the commitment to cell death. MOMP is regulated by the tight interplay of members of the Bcl-2 family of proteins, which play the central role in this crucial step. The interactions and balance between pro- and anti-apoptotic members of this family ensures the fine-tuning of life or death decisions at the outer mitochondrial membrane (OMM). Pro-survival members sequester activator BH3 proteins or executioners to prevent pore formation. Sensitizer proteins sequester and inhibit the pro-survival members and promote MOMP, thereby getting sequestered themselves to limit their impact.

Oncol Res Treat 2015;38(suppl 5):1–270

99


CONTENTS AUTHOR INDEX

Changes for the equilibria of these mutual sequestrations and inhibitions within dynamic complexes of Bcl-2 proteins define the decision for survival or death at the OMM. In our study we use fluorescence spectroscopy techniques to analyze these complex interactions of anti-apoptotic BclXL with pro-apoptotic Bax and Bid and Bad in solution, in an in vitro membrane system modeling the OMM and in isolated mitochondria. Our findings show that the interplay of pro-survival Bcl-XL with the pro-apoptotic counterparts is not limited to heterodimeric interactions but involves formation of higher order complexes that are crucial in the regulation of membrane permeabilization. As this key step of apoptosis is embedded in the mitochondrial membrane, the finding of new membrane associated complexes of Bcl-2 family proteins will extent our tools for the development of powerful therapeutics, which will target were it matters, in the membrane. Disclosure: No conflict of interest disclosed. V310

Endothelial cell derived micro-vesicular proteins induce breast cancer cell migration Ferraro D.1, Goosen R.1, Zanivan S.2, Patella F.2, Christofori G.1, Buess M.3 Department Biomedizin Universität, Basel, Switzerland, 2Beatson Institute for Cancer Research, Glasgow, United Kingdom, 3St. Claraspital, Basel, Switzerland 1

Introduction: The microenvironment is a central regulator of tumor biology. While the contribution of fibroblasts has been largely studied, the role of endothelial cells as regulators of cancer cell behavior is still poorly understood. As in a diverse spectrum of physiological processes in normal tissue, endothelial cells may exert a similar regulatory control in malignant cancer progression and metastasis, not only contributing to vessel formation, but also through endothelial cell specific signaling. Methods: To characterize the functional effects of endothelial-cancer interaction we focused on an in vitro model of SKBR3 breast cancer cells and human endothelial cells. Results: SKBR3 cells treated with HUVEC derived supernatant show significantly increased migratory potential, without a parallel increase in proliferation, an elongated phenotype and expression of mesenchymal markers (up-regulation of FN1, Stress Fibers and Focal Adhesion formation). The pro-migratory effect is significantly more pronounced when the supernatant is obtained from a sparse and highly proliferative endothelial cell culture than from confluent and resting endothelial cells. To better understand the differential regulation on cancer cell migration, we analyzed the supernatant of sparse or dense endothelial cells by quantitative MS proteomics (SILAC analysis). Interestingly, extracellular matrix proteins were enriched in dense endothelium supernatant. Amongst them, Biglycan reduced the pro-migratory effect of treatment with sparse endothelium supernatant, suggesting a potential role of resting endothelium as an inhibitor of cancer cell migration. The proteomic analysis of sparse endothelial cell supernatant revealed an enrichment in proteins (e.g. rap1, mmp1, annexinA2) belonging to the micro-vesicular compartment. Knocking down these proteins significantly reduced the pro-migratory effect of the endothelial supernatant on cancer cells, demonstrating that the microvesicular compartment can play a role in the modulation of tumor aggressiveness. Conclusions: We suggest that identification of the endothelial cell´s role in cancer progression independent from vessel formation could reveal targets for novel therapeutic strategies.

Fortbildung

Hepatische Tumore V313

Biliray tract cancer: Adjuvant treatment – pros and cons Stein A.1 Universitätsklinikum Hamburg-Eppendorf, Hubertus Wald Tumorzentrum II. Medizinische Klinik und Poliklinik, Hamburg, Germany 1

The treatment of biliary tract cancer has made relevant progress during the last years with improved understanding of disease biology (e.g. detection of proliferation and inflammation subtypes in intrahepatic cholangiocarcinoma) or development of highly active and well tolerated systemic treatment for advanced disease stages (gemcitabine and cisplatin). Despite decades of intense discussion wether and how to administer adjuvant treatment in biliary tract cancer available data are highly limited. Only two underpowered randomized trials about adjuvant chemotherapy are currently available showing either no benefit or a benefit limited to gallbladder carcinoma with a non contemporary chemotherapy-regimen containing mitomycin and 5FU. Pooled observational data point towards a benefit particularly in lymph node positive disease for chemotherapy or chemoradiation. Currently, several randomized trials have completed accrual (BILCAP: observation ± capecitabine; ACCORD18 – PRODIGE 12: observation ± gemcitabine and oxaliplatin) or are ongoing (ACTICCA 1: observation ± gemcitabine and cisplatin), which will likely solve the current dilemma about administration of adjuvant chemotherapy. Disclosure: No conflict of interest disclosed. V314

Metastatic liver disease: Review on different entities and different loco-regional approaches Wöll E.1 St. Vinzenz Krankenhaus Zams, Innere Medizin, Zams, Austria

1

Surgery of hepatic metastases is well established in colorectal cancer if complete resection can be achieved and liver function can be obtained. Long term results show a curative potential for certain patients after complete resection with clear margins. Ablative techniques and stereotactic radiotherapy can be combined with resection in some cases or can substitute for resection. Optimal indication and patient selection for the different approaches however are still unclear. Surgery has the advantage of complete histological workup including information on resection margins. Different ablation techniques and radiotherapy however are less invasive and post intervention morbidity might be lower. Even if combined with modern MRI imaging techniques however exact information on circumferential margins after ablation or radiotherapy can not be given. During the last decades surgery of liver metastases was performed not only in colorectal cancer and neuroendocrine cancer but also in melanoma, sarcoma, breast cancer, gastric cancer, and several other tumor entities. The role of local treatment of liver metastases in non colorectal, non neuroendocrine metastatic liver disease and the optimal approach, although feasible and safe, is not well defined yet. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

100

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Expertenseminar Stem Cell Biology V316

Quantitative approaches to single cell analysis: An update Etzrodt M.1 ETH Zurich, D-BSSE, Basel, Switzerland

1

Understanding the molecular processes that govern cell fates is essential to study the biology of normal and malignant hematopoiesis. This requires quantification of molecular and cellular behavior at the single-cell level, because bulk readouts can mask the inherent heterogeneity of the populations studied. Recent advances now permit high- throughput molecular readouts from single cells as well as continuous, noninvasive observation of cell behavior over time. Likewise new tools for single cell handling, sample preparation and culture, based on microfluidic technologies facilitate the execution of complex experiments with single cell precision, in an automated fashion and at reduced cost. Each of the available approaches has specific advantages and limitations. Most molecular profiling approaches for instance provide only snapshots of a given cellular state and represent end-points, which prevents to link a molecular readout to future fates of the cells investigated. The knowledge of future fates is a prerequisite though when studying how a specific molecular signature influences the differentiation of a single normal or leukemic transformed hematopoietic stem or progenitor cell (HSPC). Consequently for many questions in the field the continuous, noninvasive observation of single cell behavior over time is an absolute requirement. This seminar will address how single cell technologies, including advances in time-lapse imaging and molecular profiling, can be applied in hematology to study normal and leukemic HSPC fate. Further it will provide an interactive forum to discuss how both approaches (destructive and non-destructive) can be combined in a meaningful way to the study of normal and malignant hematopoiesis. Disclosure: No conflict of interest disclosed.

Expertenseminar

Allogene Stammzelltransplantation V317

Allogeneic stem cell transplantation – current concepts and future directions Hemmati P.1, Arnold R.1 Charité – Universitätsmedizin Berlin, Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany 1

Within the last two decades allogeneic hematopoietic stem cell transplantation (alloHSCT) has evolved into an important treatment option for a broad spectrum of malignant and non-malignant diseases of the hematopoietic system. As compared to conventional therapeutic approaches, e.g. chemotherapy, alloHSCT offers the highest potential for achieving remission and, ultimately, cure from the underlying disease. This strongly underscores the importance of alloreactivity in mediating the graft-versus-tumor effect as a prerequisite for long-term disease control. With 15.000 allogeneic transplants performed annually in Europe the use of alloHSCT has grown exponentially with respect to indications. Furthermore, the development of reduced-intensity/toxicity conditioning regimens has allowed expanding alloHSCT to elderly and/or frail patients ineligible to standard conditioning. Likewise, advances in supportive care have led to a substantial reduction in short and long-term morbidity and mortality, one of the major obstacles for achieving successful treatment outcome. Finally, improvements in HLA-typing have allowed for better donor selection.

Abstracts

Despite tremendous progress has been made in recent years, many aspects of alloSCT remain to be settled. Burning questions include: how to choose the most appropriate donor and what is the role of alternate stem cell sources? How and when to modify conditioning and post-transplant immunosuppression to optimize the balance between disease control and transplant-related toxicity? What is the optimal tool for post-transplant monitoring of minimal-residual disease? In addition to specific conditions during the intermediate post-transplant interval, e.g. acute graft-versus-host disease or infections, a substantial proportion of patients survive long-term and, therefore, are prone to a unique set of complications and late effects. This, in turn, prompts the need to develop novel follow-up strategies. These and other issues will be interactively presented and discussed during the session. Disclosure: No conflict of interest disclosed.

Fortbildung ALL V319

Molecular diagnostic in ALL: Standards and novel approaches Baldus C.D.1 Charité Campus Benjamin Franklin, Hämatologie/Onkologie/ Tumorimmunologie, Berlin, Germany 1

Acute lymphoblastic leukemia (ALL) represents a clinical and molecular highly heterogeneous disorder. Thus, a comprehensive diagnostic work up is necessary to allow risk stratification and the detection of therapeutic targets. Whereas the compulsory diagnostic program comprises standard morphology, FACS, cytogenetics, and molecular genetics, novel insights are gained by genome wide studies. The morphological assessment of lymphoblasts by microscopy is essential to confirm or rule out L3 morphology as hint for a Burkitt leukemia. The immunophenotypic determination of lineage commitment is critical for the correct diagnosis of ALL; phenotypes with therapeutic implications are T-cell, mature B-cell, B-cell precursor phenotypes. The chromosomal analysis is an integral component of the initial work-up and identifies aberrations in 60–80% of patients. In addition, highly specific and sensitive molecular techniques, such as RT-PCR, fluorescence in-situ hybridization, allow detection of fusion transcripts and chromosomal translocations with prognostic or therapeutic relevance. Importantly, t(9;22)/BCR-ABL directs the use of tyrosine kinase inhibitors (TKI). As treatment response is highly predictive of the risk of relapse, the identification of leukemia-specific markers (immunoglobulin, T-cell receptor genes, fusion transcripts) at diagnosis is necessary to enable monitoring of residual clonal ALL cells, e.g. minimal residual disease (MRD), during the course of treatment. In addition to this required diagnostic work-up, insights form exploratory studies are emerging and will shape a novel work-up in the near future. Gene-expression profiling has shown to accurately identify major ALL subtypes, and allowed the recognition of a new subtype, the Philadelphia-like ALL. By genome wide sequencing studies this novel high-risk subtype was characterized by alterations that activate cytokine receptors or tyrosine kinases amenable to inhibition with TKI. Genomic profiling has also enabled the identification of clonal heterogeneity and genetic alterations enriched at relapse. Many of these findings are of clinical relevance, thus efforts are made to capture molecular alterations in novel diagnostic tests including NGS based gene panels. A detailed molecular classification at diagnosis and during the course of ALL is highly relevant to guide therapeutic decisions for the individual patient and to allow the implementation of molecular directed therapy approaches. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

101


CONTENTS AUTHOR INDEX

Fortbildung

Melanom neue Substanzen und Strategien V324

Radiation therapy for melanoma: Adjuvant, palliative and abscopal effects Rogers S.1, Datta N.1, Bodis S.1 Kantonsspital Aarau, Radio-Onkologie-Zentrum KSA-KSB, Aarau, Switzerland

1

Radiation therapy (RT) has long played a role in the management of melanoma, however historically its efficacy has been questioned due to the perceived radioresistance of melanoma cells in vitro. Apart from proton therapy for choroidal melanoma, RT has limited accepted indications as a definitive treatment. Surgery remains the first line therapy of choice for operable patients and adjuvant RT can be considered for high-risk primary tumours. Prospective randomised data are awaited following a Phase II TROG study reporting high local control rates (93%) following postoperative nodal irradiation in selected patients with node-positive melanoma. High response rates of melanoma (e.g. mucosal or inoperable) to primary radiotherapy have also been reported. Local radiation can achieve tumour-antigen release and clinical results suggest biological interaction with immunomodulating agents (a checkpoint inhibitor and anti-CTLA4 antibody), corroborated by preclinical data. Much interest also lies in enhancing melanoma radiation response rates through combination with clinical hyperthermia, which can achieve radiosensitisation, at least in part, by immune modulation. Novel developments in radiotherapy include the rise in radiosurgical treatment (SRS), involving high dose, small volume irradiation. SRS is increasingly the palliation of choice for patients with two or more brain metastases, following resection of a single brain metastasis and for extra-cranial oligometastases. The results of a Phase III trial evaluating whole brain radiotherapy are awaited. The radiobiological consequences of SRS may be even more immunomodulatory, congruent with retrospective data that fraction sizes greater than 4 Gray are more effective and SRS alone has been suggested to induce an abscopal effect. To date there are limited data reporting SRS with concomitant immunomodulators and the optimal combinations remain to be defined. Serious toxicities have been reported following administration of B-RAF inhibitors with and subsequent to both SRS and fractionated RT. Combination strategies are being actively researched as these should increase the effectiveness of radiation in the management of patients with melanoma by widening the therapeutic window between normal tissue toxicity and melanoma cell kill, and by potentially inducing an out-offield abscopal effect. Disclosure: No conflict of interest disclosed.

e.g. breast screening including breast MRI or early detection of pancreatic cancer, 2) prophylactic surgery, particularly prophylactic salpingo-oophorectomy and 3) predictive testing of healthy family members. The knowledge of a BRCA1 or BRCA2 mutation has implications for treatment: due to the significantly increased risk of secondary breast cancer (ipsilateral or contralateral) after the first diagnosis of breast cancer, therapeutic (and prophylactic contralateral) mastectomy versus breast-conserving therapy and radiotherapy have to be discussed. Chemotherapy may be adapted because of the better response to platinum derivatives and the promising new PARP inhibitors. Fast-track genetic diagnostics can be carried out within a few days to aid decision-making. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium Pankreaskarzinom V331

Advances in the field of pancreatic cancer: What is the role of whole genome sequencing? Wicki A.1 Universitätsspital Basel, Onkologie, Basel, Switzerland

1

Next generation sequencing (NGS) is supposed to change the face of cancer care. With the help of a single sequencer, 8 complete cancer genomes can now be analysed in a single day. Thus, from a technical point of view, there are no obstacles preventing patients and physicians to look at individual cancer genomes in depth. However, the new challenge consists in interpreting NGS data and turning them into something that benefits cancer patients. With regard to pancreatic cancer, whole genome sequencing plays a double role: on one hand, whole genome sequencing can identify mutations or copy number variations of druggable oncogenes that occur in a significant proportion of pancreatic cancers, but at a low individual patient prevalence. In addition, NGS has already identified new driver genes of pancreatic carcinogenesis, and those may become actionable in the future. On the other hand, whole genome sequencing can help to assess the occurrence and structure of neo-antigens in pancreatic cancer. Together with new approaches of modulating the immune environment of a tumor, this may help to develop tailored immunotherapies for this disease. Disclosure: No conflict of interest disclosed.

Fortbildung

Urothelkarzinom – Management 2015

Fortbildung

Mammakarzinom Standards in der adjuvanten Behandlung

V334

V326

Tumors of the upper urinary tract and urethra – multidisciplinary management

Hereditary breast cancer – diagnosis, prevention, treatment

Gakis G.1

Morlot S.1, Schlegelberger B.1

1

Hannover Medical School, Institute of Human Genetics, Hannover, Germany

1

Approximately 5% of all breast cancers and more than 10% of all ovarian cancers occur as a result of a hereditary predisposition. Possible indications of a hereditary cause can be found in the family and patient’s own medical history (frequent breast, ovarian cancer, pancreatic and/or prostate cancer in the family, young age, triple-negative breast cancer, bilateral breast cancer, male breast cancer, serous ovarian cancer). In about 20–25% of these “high-risk” families, a mutation can be found in one of the known tumor predisposition genes (BRCA1, BRCA2, CHEK2 and other DNA repair genes). It is of the utmost importance to identify these “high-risk” families in order to offer 1) intensified screening measures,

102

Oncol Res Treat 2015;38(suppl 5):1–270

Klinik für Urologie, Tübingen, Germany

Introduction: The treatment of rare urogenital tumors represents a major clinical challenge. Due to their low incidence, most tumors of the upper urinary tract and urethra are diagnosed in locally advanced stages. Moreover, by contrast to upper tract urothelial carcinoma (UTUC), urethral cancers can arise from different histological entities which inherits therapeutic implications. Material and methods: Based on current data from European and North American cancer registries, improvements in the understading of the biology of tumors of the upper urinary tract and urethra have been recently made. In addition, the European Association of Urology has published this year the first update of the 2013 Guidelines on Primary Urethral Carcinoma.

Abstracts


CONTENTS AUTHOR INDEX

Results: Radical nephroureterectomy (RNU) represents the mainstay of treatment for UTUC. Yet, solitary, low-grade and low-stage upper tract tumors can be treated safely with a kidney-sparing approach, especially when located in the ureter. Some retrospective studies suggest that neoadjuvant cisplatin-based, multi-agent chemotherapy can be offered to patients with locally advanced UTUC to reduce the risk of micrometastatic disease prior to RNU. In localized urethral carcinoma, a urethral-sparing approach should be considered, if negative surgical margins can be achieved intraoperatively. In locally advanced urethral cancer, cisplatin-based multiagent chemotherapy (or a radiosensitizing chemotherapy with concurrent radiotherapy for patients with squamous cell carcinoma) may exert a beneficial impact on survival and even enable genital preservation. In case of urethral recurrence, recent data suggest that salvage surgery is associated with an improved overall survival. Conclusions: Low-stage and low-grade tumors of the upper urinar tract and urethra can be safely treated with organ-sparing surgery, whereas a multidisciplinary approach should be especially considered for patients with locally advanced tumors. Disclosure: No conflict of interest disclosed.

Fortbildung

Psychoonkologie V339

“Cancer-related fatigue: What is to do?” Fischer I.1,2 Institut für Tumor-Fatigue-Forschung, Emskirchen, Germany, 2Deutsche Fatigue Gesellschaft, Köln, Germany 1

Cancer-related fatigue (CrF) is defined as a persistent, unusual , subjective sensation of physical, emotional and cognitive tiredness which occurs in the context of cancer or its treatment. Compared with the tiredness felt by a healthy individual, CrF is perceived as being of greater magnitude, disproportionate to recent activity, and it is not completely relieved by rest, leaving the patient with an overwhelming and distressing sense of exhaustion. Prevalence estimates of CrF range from 20% to 99%, depending on the sample, the time and the method of assessment. The CrF symptoms can be temporary, but can also persist up to 15 years after successful treatment completion. Having a profound effect on the patient´s life, CrF is much more than just being tired: Patients are often unable to engage in their usual activities of daily living (up to the inability to perform one’s job and earn a living), social relationships are lost and quality of life is impaired. Thus, CrF clearly poses a problem not only for the patients themselves, but also for the persons around them. Although evidence-based therapies for CrF are available and though the National Comprehensive Cancer Network (NCCN, 2015) recommends that CrF should be “recognized, evaluated, monitored, documented and treated promptly for all age groups, at all stages of disease, prior to, during, and following treatment”, CrF is not sufficiently perceived by physicians: Individuals are often reluctant to report fatigue, and cancer-care providers frequently do not screen for it because they are uncertain about how to treat it. However, patients are suffering, and since fatigue is related not only to cancer, but also to other diseases and symptoms (e.g. underlying comorbidities, emotional distress, anemia, sleep disturbance, pain, or adverse events of medication ), diagnosis and differential diagnosis are a significant precondition for an adequate causal and/ or symptomatic treatment of CrF. This further education provides an overview about adequate screening methods and diagnostic procedures (e.g. anamnesis, suitable self report questionnaires) as well as about symptomatic treatment options supported by evidence from systematic reviews, meta-analyses and randomized controlled clinical trials (the latter with CrF as a primary endpoint), such as physical training, patient education, cognitive behavior therapy, energy conservation, phytotherapy, (psycho-)stimulants or corticosteroids.

Freier Vortrag Stammzellen I V341

Effective mobilization of hematopoietic stem cells with a small molecule α9β1/α4β1 integrin antagonist Grassinger J.1, Cao B.2,3, Zhang Z.2, Klatt S.1, Williams B.2,3, Mueller G.1, Hart C.1, Schelker R.1, Herr W.1, Nilsson S.K.2,3 University Hospital Regensburg, Internal Medicine III, Regensburg, Germany, 2Commonwealth Scientific and Industrial Research Organization, Manufacturing Flagship, Melbourne, Australia, 3Monash University, Australian Regenerative Medicine Institute, Melbourne, Australia 1

Introduction: Mobilization of sufficient numbers of hematopoietic stem cells (HSC) is a prerequisite for a successful engraftment in peripheral blood stem cell transplantation (PBSCT). To date, granulocyte-colony stimulating factor (G-CSF) is the most effective mobilization agent used clinically. However, G-CSF has substantial nonspecific effects on peripheral blood (PB) and bone marrow (BM) cells and, according to our data, mainly expands hematopoietic stem and progenitor cells (HSPC) within the central BM region. We recently showed that biologically superior HSC are located at the endosteal BM region and therefore propose that mobilization of these HSC will result in superior engraftment than G-CSF mobilized HSC. Methods: A small molecule α9β1/α4β1 integrin antagonist (N-(Benzene-sulfonyl)-L-prolyl-L-O-(1-Pyrrolidinylcarbonyl)-tyrosine; BOP) and the fluorescent analogue (R-BC154) was synthesized and shown to disrupt the binding of HSC to extracellular ligands within the endosteal niche. In vitro and in vivo analysis was performed to investigate the binding capacity of BOP and HSC mobilization efficiency. Results: Using R-BC154, we show that this class of antagonists preferentially bind mouse and human HSC via intrinsically activated α9β1/α4β1 integrins within the endosteal niche. BOP rapidly mobilizes HSC with long-term multi-lineage engraftment potential in mice. Additive augmentation of the engraftment of PB-HSC was observed when BOP was co-administered with a SDF-1 antagonist (Plerixafor). Impressively, this combination effectively outcompeted PB-HSC mobilized with 4 days of G-CSF treatment. The enhanced mobilization observed with the small molecule combination was recapitulated in humanized NSG mice, where a significant increase in PB CD34+ HSPC was observed after treatment with BOP and Plerixafor. Conclusions: Dual α9β1/α4β1 integrin inhibitors effectively mobilize murine and human HSC in mice. The combination of BOP and Plerixafor might allow efficient single dose mobilization strategies with reduced side effects as compared to multiple G-CSF injections. Moreover, mobilization of aberrant hematopoietic cells in acute and chronic myeloid leukemia using BOP might enhance treatment efficiency and is currently under investigation. Disclosure: No conflict of interest disclosed. V342

The role of the transcription factor trps1 an its relation to evi1 in zebrafish hematopoietic development Alghisi E.1, Konantz M.1, Lengerke C.1,2 University Hospital Basel, Department of Biomedicine, Basel, Switzerland, University Hospital Basel, Clinic for Hematology, Basel, Switzerland

1 2

Disclosure: No conflict of interest disclosed.

Introduction: The EVI1 (ecotropic viral integration site 1) gene is a zinc finger transcriptional regulator that is expressed in hematopoietic stem cells (HSC) and our lab has shown that it regulates their emergence during development via up-regulation of the NOTCH pathway in the aorta-gonado-mesonephros (AGM) region However, the precise molecular mechanism of EVI1-mediated NOTCH-induction is unclear (Konantz et al., unpublished). Here we study the trichorhinophalangeal syndrome 1 (trps1) gene, a GATA family transcriptional factor known for its role in skeletal

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

103


CONTENTS AUTHOR INDEX

development, with respect to its role in evi1-mediated hematopoiesis. TRPS1 has been reported to induce epithelial-to-mesenchymal transition and angiogenesis in breast carcinoma (BC) cells through activation of its direct target gene VEGFA. Previous data from our lab report that VEGF overexpression can rescue HSC defects in evi1-morphants. Methods: Genetic modifications were obtained by injection of inhibiting morpholino oligonucleotides or mRNA into the ZF zygote or via heatshock inducible transgenic lines. Hematopoiesis was analysed by in situ hybridization for trps1, runx1, cmyb, rag1, notch1b and notch3 in the AGM, the caudal hematopoietic tissue (CHT) and the thymus. Results: Zebrafish embryos show trps1 expression in the brain, the jaw mesenchyme and potentially in the AGM, and later in development also in the kidney marrow. Knockdown of trps1 reduced the formation of cmyb/runx1-double positive HSC in the AGM region as well as the formation of rag1 positive lymphoid cells in the thymus, indicating a functional role in definitive hematopoiesis. Importantly, trps1 morphants showed down-regulation of NOTCH pathway genes in the dorsal aorta and AGM regions suggesting an upstream role of trps1 in the vegf-notch axis in developmental hematopoiesis. Co-injection of evi1 mRNA was not able to rescue runx1 expression and morphants showed a normal evi1 expression compared to controls, indicating that indeed trps1 may act as an evi1 downstream target. Rescue experiments of evi1 morphants with trps1 mRNA further exploring this hypothesis are underway. Conclusion: Taken together, our data indicate an unsuspected role of trps1 in hematopoiesis and suggest this transcription factor as a possible downstream target of evi1 during blood stem cell development. We are currently exploring trps1 as a direct target of evi1. Disclosure: No conflict of interest disclosed. V343

Wnt5a expressed by the niche maintains migratory properties of hematopoietic stem cells through the planar cell polarity pathway Schreck C.1, Istvanffy R.1, Ziegenhain C.2, Gärtner F.3, Grziwok S.1, Pagel C.1, Henkel L.4, Schiemann M.4, Götze K.1, Massberg S.3, Peschel C.1, Enard W.2, Oostendorp R.A.J.1 III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany, 2Department of Biology II, Ludwig-MaximiliansUniversität, Anthropology and Human Genetics, München, Germany, 3LudwigMaximilians-Universität, I. Medizinische Klinik, München, Germany, 4Technische Universität München, Institute of Medical Microbiology, Immunology, and Hygiene, München, Germany 1

We previously identified Wnt5a as a secreted factor that maintains HSCs in vitro (Buckley, et al., Exp Hematol. 2011) and as a factor responsible for decline of HSC function during aging (Florian et al., Nature. 2013). We here studied in detail how Wnt5a-deficiency affects the niche and its ability to maintain long-term repopulating HSC. The BM niche in Wnt5a-deficient mice show increased number of CD31+ endothelial cells. The number of mature and immature mesenchymal cells was not different, but multipotent stromal cells (MSCs) showed increased proliferation and calcification upon differentiation. Chimeric transplantation experiments showed that Wnt5a± HSCs engraft similarly to wild-type Wnt5a+/+ (WT) HSCs up to tertiary transplantations. To assess whether niche Wnt5a affects engraftment, and/or self-renewal capacity of HSCs, wild-type (WT) HSCs were transplanted in Wnt5a-deficient recipients. We found that in primary recipients, engraftment was similar to that in WT recipients. Unexpectedly, however, WT donor LSK cells from the Wnt5a-deficient recipients completely failed to engraft in secondary recipients. Although the phenotype of the WT donor cells engrafted in either WT or Wnt5a± environments was similar, the sorted LSK cells from Wnt5a± recipients showed defective, apolar distribution of Cdc42 and F-actin, suggesting possible defects in migratory properties. Indeed, RNAseq of sorted donor LSK cells from Wnt5a± recipients showed highly divergent expression of molecules involved in heterotrimeric G-protein assembly and small GTPase-mediated planar cell polarity (PCP) signal transduction. Further

104

Oncol Res Treat 2015;38(suppl 5):1–270

experiments showed that WT donor LSK cells from Wnt5a± recipients showed defective lamellipodia formation, and only poorly homed to the bone marrow of recipient WT mice. In addition, mature T and B lymphocytes failed to migrate towards CXCL12. In conclusion, our experiments show that a Wnt5a-deficient niche allows for initial engraftment, but fails to maintain the ability of engrafted HSCs to properly home and engraft secondary recipients, due to defective PCP-regulated migratory properties of HSCs and mature lymphoid cells. Disclosure: No conflict of interest disclosed. V344

Allogeneic T cells disrupt medullary thymic epithelial cell formation and indirectly lead to chronic graft-vs-host disease Müller A.M.1,2, Florek M.2, Min D.3, Burnett C.2, Weinberg K.3, Shizuru J.A.2 UniSpital Zürich, Hämatologie, Zürich, Switzerland, 2Stanford University, BMT Medicine, Stanford, United States, 3Stanford University, Stanford, United States 1

Graft-vs-host disease (GVHD) remains a major complication of allogeneic hematopoietic cell transplantation (HCT). Acute (a) GVHD results from direct donor T cell (TC) damage of organs. In contrast, the biology of chronic (c) GVHD with its various autoimmune-like manifestations remains poorly understood. We studied the pathophysiology of cGVHD in an MHC-matched mouse model (C57BL/6 à BALB.B) using lethal irradiation and HCT of pure hematopoietic stem cells (HSC; cKIT+Thy1.1loSca1+Lin-) or HSC + TC. Recipients of pure HSC remained healthy, whereas mice given HSC+TC developed aGVHD (diarrhea, weight loss) with a mortality of ~30%. Survivors stabilized around d+45, but developed clinical chronic GVHD after 6–12m with sclerodermatous skin excoriations, cataracts, and liver fibrosis/cirrhosis. Further, thymuses were a major target of aGVHD resulting in severe hypocellularity and disrupted organ architecture. During the months of lymphoid reconstitution thymuses of HSC+TC-recipients showed markedly lower expression of cytokeratin 5 (CK5) than HSC recipients. CK5 marks medullary thymic epithelial cells (mTEC) that provide a specialized microenvironment for survival, proliferation, and differentiation of immature TC. Expression of AIRE, a transcription factor in the thymic medulla that controls negative selection during TC maturation was low in all transplanted groups. During aGVHD only TH1-donor TC, but not CD4+IL17+ (TH17)-cells were detectable. Starting at 2m TH17 cells emerged, first in intestines then liver and skin (all typical GVHD target organs) increasing until 6–12 months post-HCT. Of note, TH17 cells originated from donor-HSC, not adaptively transferred mature TC. Even recipients of pure HSC showed increasing proportions of TH17 cells over time, and could manifest discrete signs of cGVHD. From our model we hypothesize that the thymus is damaged by transplant conditioning and alloreactive donor T cells. Disruption of medullary thymic epithelial cells (mTEC) may lead to impaired restoration of the epithelial network. As a consequence, negative selection of nascent HSC-derived donor T cells is compromised and emergence of IL-17 secreting donor T cells in target organs of chronic GVHD could be due to misguided T-cell maturation in a disrupted thymic microenvironment. Subclinical GVH-reactions within the thymus could thereby contribute to chronic GVHD in an autoreactive fashion. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V345

Specific transcripts of DNMT3A modulate differentiation of hematopoietic progenitor cells Božić T.1, Frobel J.1, Raić A.1, Goecke T.2, Jost E.3, Wagner W.1 Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, Aachen, Germany, 2RWTH Aachen University Hospital, Department of Obstetrics and Gynecology, Aachen, Germany, 3RWTH Aachen University Hospital, Clinic for Oncology, Hematology, and Stem Cell Transplantation, Aachen, Germany 1

Introduction: DNA-methyltransferase 3A (DNMT3A) is a de novo DNA-methyltransferase that is alternatively spliced in a tissue- and disease-specific manner, but the functional relevance of these transcripts is hardly known. DNMT3A is frequently mutated in patients with acute myeloid leukemia (AML) and we have recently demonstrated that mutations in DNMT3A can be mimicked by aberrant hypermethylation within the DNMT3A sequence: about 40% of AML patients carry this “epimutation” that is associated with shorter overall survival. Notably, DNMT3A mutations as well as its “epimutations” seem to have impact on the expression of different DNMT3A transcripts. Therefore, we aimed for a better understanding of the functional role of individual DNMT3A splice variants in hematopoiesis. Methods: Specific DNMT3A transcripts (transcripts 1+3, transcript 2 or transcript 4) were knocked down by lentiviral expression of individual short-hairpin RNAs (shRNAs) in CD34+ progenitor cells isolated from human cord blood. Subsequently, we evaluated the impact on colony formation potential (CFU assay), proliferation (CFSE assay), and the immunophenotype (CD34+ and CD133+). DNA methylation profiles of individual knockdowns were generated with the Infinium HumanMethylation450 BeadChip platform and are under current analysis. Results: Knockdown efficiency of DNMT3A transcripts 1+3, transcript 2, and transcript 4 was validated by qRT-PCR (30%, 55% and 70% of basal expression, respectively). Downregulation of either transcript 2 or 4 reduced the proliferation rate significantly (N = 3, p < 0.05). Notably, cells maintained a more primitive phenotype expressing CD34+ for more cell divisions upon knockdown of transcript 2 (N = 3; p < 0.05), whereas CD133+ expression continuously declined in all knockdowns and the control. Downregulation of transcript 4 had significant effect on the CFU potential, leading to a bias for erythroid colonies, together with an increase in the total number of colonies. Conclusions: Our results indicate that individual DNMT3A transcripts have unique regulatory functions during the differentiation process of blood progenitors that might be relevant for development of AML. Disclosure: Tanja Božić: No conflict of interest disclosed. Wolfgang Wagner: Employment or Leadership Position: Wolfgang Wagner is involved in the company Cygenia that provides service for analysis of the DNMT3A epimutation (www.cygenia.com).; Honoraria: RWTH Aachen has applied for a patent for the DNMT3A epimutation. V346

Signal transduction and functional effects of G proteincoupled receptors in hematopoietic stem- and progenitor cells Manz P.1, Krauß U.1, Kanz L.1, Möhle R.1 Med. Univ.-Klinik II, Tübingen, Germany

1

Introduction: G protein-coupled receptors (GPCR), e.g. the chemokine receptor CXCR4, the leukotriene receptor CysLT1, and the sphingolipid receptor S1P1, represent important regulators of the “stem cell niche,” which supports maintenance and survival of hematopoietic stem and progenitor cells (HPC) in the bone marrow microenvironment. However, the differential effects of these GPCR and their individual roles are poorly understood. Methods: We analyzed signal transduction pathways of CXCR4, CysLT1, and S1P1 in CD34+ HPC and cell lines (THP-1), particularly calcium signaling by flow cytometry and MAP-kinase/Erk phosphorylation by

Abstracts

western blot and flow cytometry. Adhesion of HPC to stromal cells and fibronectin was analayzed in adhesion assays. Proliferation of HPC with and without coculture with stromal cells was also assessed. Results: We found that after stimulation with their respective ligands, CysLT1 induced the strongest response in calcium signaling, while CXCR4 and particulary S1P1 only moderately mediated intracellular calcium release in HPC. Using a quantitative flow cytometric assay, strong MAPK phosphorylation was mediated by CXCR4, while a modest increase was induced by CysLT1, and only a weak response observed after stimulation of S1P1. Stimulation of both CXCR4 and CysLT1 synergistically increased MAPK phosphorylation, which was not further augmented by activation of S1P1. However, only combined inhibition of all three GPCR resulted in reduced proliferation of HPC in vitro. Interestingly, a converse effect on HPC adhesion was induced by S1P1, which inhibited adhesion of CD34+ HPC to fibronectin, compared to CXCR4 and CysLT1, which both mediated an increased HPC attachment. Conclusions: We conclude that the G protein-coupled receptors CXCR4, CysLT1, and S1P1 induce differential signaling and functional effects in HPC. While CXCR4 elicited the most prominent effect on calcium signaling, MAPK phosphorylation and signaling related to cell proliferation was increased particularly by CXCR4. Our results suggest that CXCR4 and CysLT1 support lodging and proliferation of HPC in the hematopoietic microenvironment, while only weak effects of S1P1 on calcium and MAPK signaling and even reduced adhesion of HPC fits to the recently recognized opposite role of S1P1 supporting HPC egress and mobilization. Thus, different GPCR play diverse roles in the dynamic regulation of the HPC niche. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Nicht maligne Hämatologie V350

Data from German centers in the global PNH Patient Registryanalysis of the translation of the DGHO-guidelines for the treatment of Paroxysmal Nocturnal Hemoglobinuria (PNH) in real life therapy Höchsmann B.1, Leichtle R.1, Röth A.2, Panse J.3, Haferlach T.4, Borchmann P.5, Aulitzky W.E.6, Dengler J.7, Port M.8, Platzbecker U.9, Klausmann M.10, Steinmetz T.11, Becker M.12, Schmidt B.13, Schrezenmeier H.1 Institut für klinische Transfusionsmedizin und Immunogenetik Ulm, DRK Blutspendedienst Baden-Württemberg-Hessen und Universität Ulm, Ulm, Germany, 2Universitätklinik Essen, Abteilung Hämatologie, Essen, Germany, 3 Universitätsklinikum Aachen, Klinik für Onkologie, Hämatologie und Stammzelltransplantation (Medizinische Klinik IV), Aachen, Germany, 4MLL Münchner Leukämielabor GmbH, München, Germany, 5Universitätsklinikum Köln, Klinik I für Innere Medizin, Köln, Germany, 6Robert Bosch Krankenhaus Stuttgart, Hämatologie und Onkologie, Stuttgart, Germany, 7Medizinische Universitätsklinik Heidelberg, Abteilung Innere Medizin V, Heidelberg, Germany, 8 Medizinische Hochschule, Abteilung Hämatologie und Onkologie, Hannover, Germany, 9Universitätsklinik Dresden, Abteilung Hämatologie und Onkologie, Dresden, Germany, 10Gemeinschaftspraxis Drs Klausmann und Dr Welslau, Aschaffenburg, Germany, 11Gemeinschaftspraxis Hämatologie und Onkologie, Köln, Germany, 12Onkologische Praxis Minden/Porta, Minden, Germany, 13 Onkologische Praxis Pasing, München-Pasing, Germany 1

Introduction: The International PNH Patient Registry was started to enhance the understanding of this orphan disease and includes patients (pts) independent of disease severity or treatment. Complement inhibition with eculizumab is capable to improve the clinical symptoms and overall survival of PNH pts. An expert panel of the DGHO defined guidelines for diagnosis and treatment of PNH (https://www.dgho-onkopedia.de). According to recent data confirming a strong correlation between clinical symptoms and prognosis eculizumab is recommended for symptomatic pts with hemolytic PNH.

Oncol Res Treat 2015;38(suppl 5):1–270

105


CONTENTS AUTHOR INDEX

Tab. 1. zu V350 PNH related symptoms and treatment status

Physician reported Symptoms and medical events

All Patients at Baseline in% (n = available pts)

Never Treated with Eculizumab, at Baseline, Patients in%, (n = available pts)

Ever Treated with Eculizumab, at Baseline, Patients in%, (n = available pts)

All Patients with Worsening from Baseline to Last Follow-Up (FU), in%, (n = available pts)

Ever-Treated with Eculizumab with Worsening from Baseline to Last FU, Patients in%, (n = available pts)

Never Treated with Eculizumab with Worsening from Baseline to Last FU, Patients in%, (n = available pts)

Any thrombotic event

18.0% (n = 345)

12.1% (n = 140)

22.0% (n = 205)

2.7% (n = 187)

1.2% (n = 85)

3.9% (n = 102)

Abdominal Pain

35.5% (n = 220)

30.2% (n = 129)

48.4% (n = 91)

10.6% (n = 151)

6.2% (n = 70)

14.5% (n = 81)

Hemoglobinuria

50.5% (n = 218)

41.9% (n = 129)

70.8% (n = 89)

17.9% (n = 140)

12.7% (n = 63)

22.1% (n = 77)

Methods: To study the adherence to the DGHO guidelines we analysed the rate of symptomatic PNH pts in the PNH Patient Registry which collects pseudonymized patient data after obtaining an informed consent. Data were collected at baseline and the last follow up (FU) in all pts and in the subgroups of pts ever or never treated with eculizumab. Furthermore, the worsening of the disease defined as the new occurrence of symptoms from baseline to the last FU was investigated. Baseline is defined as the start date of eculizumab for treated pts or registry enrollment date for never treated pts. Last FU is defined by the first reported date of death, BMT, withdrawal from the Registry, or discontinuation of eculizumab (in treated pts). In pts not meeting any of these criteria, last FU is the date of last contact from the Registry. Results: At enrollment the mean age was 45.1 ± 18.42 yrs, mean GPI-deficient granulocyte population was 52.8 ± 34.32, mean LDH was 3.9 ± 3.55 xULN, The mean follow up duration was 2.2 ± 2.2 yrs in the complete pts group, 2.5 ± 2.3 yrs in the pts ever treated with eculizumab and 1.9 ± 2.1 yrs in the pts never treated with eculizumab. The table below shows the data of the PNH related symptoms according to the treatment with complement inhibition. Conclusion: A substantial subgroup is not treated with eculizumab despite symptoms. Status deterioration, including new thromboembolic events, was more pronounced in the subgroup never treated with eculizumab. We will present further analyses of the German PNH Patient Registry regarding combination of clinical signs and therapeutic decisions to enable an further improvement of the treatment strategy in symptomatic PNH. Disclosure: Britta Höchsmann: Advisory Role: ja; Financing of Scientific Research: ja; Expert Testimony: ja Hubert Schrezenmeier: Advisory Role: ja; Financing of Scientific Research: ja; Expert Testimony: ja.

Lysophosphatidic acid (LPA) induces a concentration dependent calcium influx in RBCs. We found that this influx happens via a bifurcated signal pathway. The signal transduction starts with the common LPA-receptor agonist binding and the consecutive activation of the Gi-Protein, which triggers the two branches: (i) activation of MEK/MAPK and PI3 kinase followed by an ω-Agatoxin TK sensitive calcium-entry and (ii) activation of the non selective cation channel TRPC6. In detail the LPA induced TRPC6 cascade is: LPA -> Gi -> PLCε -> PIP2 -> DAG -> PKCα -> TRPC6. However, it was unclear, how PKC phosphorylation can activate TRPC6. We could generate evidence that the phosphorylation of TRPC6 by PKCα initially leads to an inactivation of the channel. This inactivation enables TRPC6 channels to form an assembly platform with FK binding protein and calcineurin. The dephosphorylation of TRPC6 by calcineurin then activates TRPC6 and such enables an influx of calcium into the RBC. Furthermore we could show that the function of this pathway is increased in RBCs of sickle cell disease patients through a higher abundance of LPA-receptor 4 in sickle cells. The LPA signaling pathway and the resulting activation of channels place them into the focus as pharmaceutical targets for the treatment of severe symptoms of anaemia, such as the vaso-occlusive crisis in sickle cell disease. This holds particularly true, as calcium has been identified as a trigger for RBCs self aggregation and for their adhesion to endothelial cells. Disclosure: No conflict of interest disclosed. V352

Artesunate therapy for vaso-occlusive crisis with multi-organ failure due to severe plasmodium falciparum infection in a patient with HbSC trait Trummer A.1, Thol F.1, Ganser A.1 Hannover Medical School, Hematology, Hemostasis, Oncology & Stem Cell Transplantation, Hannover, Germany 1

V351

Red cell calcium-channelopathies – novel approaches and mechanism Kaestner L.1, Wang J.1, Hertz L.1, Ruppenthal S.2, Lipp P.2, Birnbaumer L.3, Freichel M.4 Research Centre for Molecular Imaging and Screening, Homburg/Saar, Germany, 2Institute for Molecular Cell Biology, Homburg/Saar, Germany, 3 National Institute of Environmental Health Sciences, Research Triangle Park, United States, 4Institute for Pharmacology, Heidelberg, Germany 1

The molecular origin of hereditary anaemia is sometimes known but often obscure. However, in both situations, the molecular mechanism leading to the symptoms of the disease remain in many cases elusive. We present single-cell based methods that allow the investigation of functional properties of red blood cells (RBCs) upon exposure to pharmacological and physical stimulation. We investigated in particular the molecular basis of calcium entry by video imaging of intracellular calcium combining pharmacological inhibitors and transgenic animal models (mice).

106

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: Hemoglobin sickle cell disease (HbSC) is the second most frequent hemoglobinopathy after homozygous sickle cell anemia (SCA). Its clinical course often differs from SCA as it is mostly complicated by thrombotic complications due to hyperviscosity rather than hemolytic-related vasculopathy. Like SCA, HbSC has been associated with a significant risk reduction of malaria. Nevertheless, malaria chemoprophylaxis is often recommended as malaria can be a significant cause of morbidity and mortality in sickle cell patients. Case: We report the case of 45 year old west-african man with HbSC trait who had only been transfused with RBC once before. He had initially reported to our outpatient clinic due to ongoing bone pain for about 6 months. However, his diagnostic work-up showed only mild hemolysis and no signs of osteonecrosis. Hemoglobin levels were around 14 g/dl with a reticulocytosis of 170/nl, and hemoglobin electrophoresis revealed 51% HbS, 46% HbC and 3% HbA. As the patient refused phlebotomy, we initiated a therapy with hydroxyurea (500mg/day) and analgetics, which led to a significant clinical improvement. However, the patient left Germany for a 3 month stay in Togo,

Abstracts


CONTENTS AUTHOR INDEX

taking chloroquine for malaria prophylaxis. Again, he developed severe bone pain during the last 3 weeks of his stay and, finally, he reported to our department having fever with chills. Ebola infection was ruled out, but the patient worsened dramatically and rapidly developed multi-organ failure with massive haemolysis, liver cell necrosis, acute kidney failure requiring haemodialysis, paralytic ileus, osteonecrosis and acute myocardial infarction. A thick blood smear revealed infection with Plasmodium falciparum (about 2‰) and a therapy with atovaquone/proguanil as well as RBC exchange transfusions was initiated. However, as the blood smear remained positive after 48 hours, therapy was switched to artesunate/doxycycline and the blood smear was found negative after another 48 hours. The patient slowly recovered and, more than 6 months later, almost all organ functions returned to normal. Conclusions: Despite a significant risk reduction in HbSC, malaria infection can cause life-threatening hemolysis and vaso-occlusive disease and clinicians need to be aware that even chemoprophylaxis does not ensure sufficient protection. Artesunate/doxycycline appears to be a rapid and efficient therapy for patients irresponsive to other malaria treatments. Disclosure: No conflict of interest disclosed. V353

Increasing number of patients with hemoglobinopathies in the Ruhr metropolitan area Distelmaier L.1, Dickerhoff R.2, Dührsen U.1 University Hospital Essen, Department of Hematology, Essen, Germany, University Hospital Duesseldorf, Department of Pediatric Oncology, Hematology and Clinical Immunology, Duesseldorf, Germany 1 2

Introduction: Due to migration hemoglobinopathies are becoming more frequent in Germany. Especially in industrial areas the number of patients with thalassemia syndromes and sickle cell disorders is increasing while there is a lack of experienced physicians. This leads to incorrect diagnosis and treatment. Methods: We summarize the numbers of patients we have seen in our hemoglobinopathy clinic at the university hospital in Essen since 2011 and evaluate the patients´ histories and former treatments. The term “patients” is used for all individuals referred to us, both asymptomatic heterozygous and patients with clinical disease. Results: There has been a significant increase of patients with hemoglobin disorders from 2011 to 2015. Starting with 5 patients in the beginning of 2011, the clinic increased to 19 patients by the end of 2012, 41 in 2013, 71 in 2014 and by April 2015 it reached a total number of 95 patients. 52 of the patients were gene carriers of either HbS, thalassemia-syndromes or hemoglobins such as HbE, HbC and others, while 42 patients had a clinical significant hemoglobinopathy that needed treatment. Within the group of significant hemoglobinopathies, sickle cell disease was the most frequent disorder with 25 patients. Furthermore we found that 45 of the 95 patients had received incorrect treatments before they came to our clinic. 15 beta-thalassemia gene carriers where erroneously diagnosed as iron deficiency and received multiple treatments with iron. 6 HbS gene carriers received treatments meant for homozygous sickle cell disease. At least 10 patients with sickle cell disease had complications related to incorrect treatments. 7 patients with beta-thalassemia major or intermedia suffered from ineffective erythropoiesis or iron overload due to an insufficient transfusion program or iron chelation. The other 7 patients were a heterogeneous group that either came with a wrong diagnosis or had received various not indicated treatments. Conclusions: Since 2011 the number of patients with hemoglobin disorders and hemoglobinopathy gene carriers that presented in our clinic has constantly increased. In Germany few physicians are experienced in recognizing and treating adults with hemoglobin disorders. For this reason many grown-up patients are still treated in pediatric clinics or are lost altogether. There is a strong need for more awareness and further education of physicians to avoid misdiagnoses and treatment-related complications. Disclosure: No conflict of interest disclosed.

Abstracts

V354

Claudia Pechstein’s doping ban – reanalysis of the blood counts which led to the ban Gassmann W.1 St. Marienkrankenhaus, Siegen, Germany

1

Claudia Pechstein has been found guilty of blood doping on the basis of increased reticulocyte counts at World Championships in Hamar in 2009 and suspicious counts in the years before. Blood counts from 2000 to 2009: This analysis comprises all 95 blood counts of Claudia Pechstein taken by anti-doping officials from 2000 to 2009. Repeatedly high reticulocyte counts may be the result of repeated use erythropoiesis stimulating agents or the result of shortened erythrocytic survival time. Aside from high retics, high MCHC values were observed in Mrs. Pechstein at many occasions suggesting an erythrocytic abnormality. Mean value was 35.7 g/dl (normal range up to 36) with values above 37 g/dl at eleven occasions. In case of blood doping, one would expect high haemoglobin levels at top events. However, mean haemoglobin levels and retics were the same at championships (mean 14.6 g/dl, 2.1%) compared to world cup competitions (14.5 g/dl, 1.9%) and compared to tests taken during training phases (14.6 g/dl, 2.0%). The haemoglobin concentration should increase about 1 g/dl or more in case doping with the use of erythropoiesis stimulating agents was the reason for the elevated reticulocytes. This has never been observed. The situation at Hamar in February 2009: Mrs. Pechstein had had five doping controls in January 2009. The first three controls were performed at the European Championship in Heerenveen. She won that championship with haematocrit levels of 39, 40 and 39% showing reticulocyte counts of 1.7%, 2.2% and 2.0%. Further checks were done at a world cup race on January 30th and 31st with haematocrit levels as in Heerenveen and reticulocyte counts of 2.4% and 2.2%. The first doping test at the Hamar championship was done on February 6th and showed a very high reticulocyte count of 3.5%. This count was confirmed at the following day immediately after a 3000m race. Haematocrit levels were determined as 41%, 39%, and 37%. Another high reticulocyte at another championship in 2011: A high reticulocyte count of 3.8% and 3.1%, respectively, was observed at the world championship at Inzell in 2011. Doping controls including blood counts had been done at seven of the last ten days prior to that high reticulocyte count. No evidence of doping was found in the samples of blood and urine showing that high reticulocyte counts can occur in Mrs. Pechstein without doping. The data available do not and did never support the suggestion of blood doping. Disclosure: No conflict of interest disclosed.

Expertenseminar

Porphyrie / Stoffwechselkrankheit V359

Acute porphyrias – diagnosis and therapy Petrides P.E.1, Bronisch O.1, Beykirch M.1 Europäisches Porphyriezentrum, Hämatologisch onkologische Schwerpunktpraxis, München, Germany 1

Several errors of porphyrin metabolism can cause the acute porphyrias, which are characterized by a 50% reduction of the activity of particular enzymes of heme biosynthesis. Under normal conditions supply of porphyrins is sufficient in spite of this impairment. However, when higher amounts are required as for instance through the increased production of heme containing cytochrome P450 enzymes, the bottleneck of metabo-

Oncol Res Treat 2015;38(suppl 5):1–270

107


CONTENTS AUTHOR INDEX

lism leads to a flood of porphyrin precursors (delta-aminolaevulinic acid (dALA) and porphobilinogen (PBG)) in the body. These precursors are the toxic molecules, which cause the acute attack and are therefore also of prime diagnostic relevance. Cytochrome P450 enzymes are produced in the liver under the influence of certain drugs, so that drugs and also hormones are the most important (if, however, not the only ones) inducers of an acute attack. The acute attack is often characterized by the symptom triad of abdominal pain, cerebral symptoms and a peripheral neuropathy. A red discoloration of the urine – in particular after extended storage – occurs in 30% of the patients and can lead to the correct diagnosis. Severe attacks – often accompanied by a hyponatremia – can cause tetraplegias with the necessity of intubation and also lead to coma. When epileptic seizures occur, MRI analysis shows PRES (posterior reversible encelopathy syndrome), i.e. a vasogenic edema in the posterior cerebral hemispheric region. When an acute porphyria is suspected the determination of PBG and dALA in a spontaneous urine sample is mandatory. When the values are elevated the presence of an acute porphyria is very likely. Further diagnosis is carried out by plasma fluorescence peak analysis, PBG-deaminase activity determination in red blood cells (in acute intermittent porphyrias) and fecal porphyrin analysis. Final proof is obtained through the identification of the disease causing mutation (genetic test). Therapy of choice is the identification of toxic substances such as drugs and their immediate discontinuation, the intravenous application of high dose glucose and – more efficiently – the application of heme arginate which is available for intravenous infusion. In this seminar, the relevant diagnostic and therapeutic aspects of the acute porphyrias will be illustrated using various examples from our cohort of nearly 50 patients with acute porphyrias which we have taken care of for the last 15 years. Disclosure: No conflict of interest disclosed.

Expertenseminar Magenkarzinom V360

Standard treatment of gastric cancer – cases discussions Lordick F.1 Universitäres Krebszentrum Leipzig (UCCL), Leipzig, Germany

1

According to the International Agency for Research in Cancer (http:// globocan.iarc.fr/Default.aspx), gastric cancer continues to be the fifth most common form of cancer from worldwide: 952,000 new patients are diagnosed with gastric cancer / year and 723,000 people die from gastric cancer. The Eurocare-5 study showed that the mean age-standardized 5-year relative survival for adult patients with gastric cancer diagnosed in 2000–2007 was only 25%, making it one of the most aggressive cancers in Europe (de Angelis, Lancet Oncol 2014). In this Expert Seminar we will discuss different disease scenarios which are typical for multimodal and potentially curative but also for palliative treatment decisions. The five cases to be discussed are: 1. 44-year-old woman with localized signet ring cell cancer of the stomach corpus, staged uT3 N0 M0. What are the next steps: Laparoscopy? Primary resection? Perioperative chemotherapy? 2. 69-year-old man with localized adenocarcinoma at the esophago-gastric junction, intestinal subtype (Lauren), uT3 N+ M0: What are the next steps: PET-CT? Primary resection? Perioperative chemotherapy? Neoadjuvant radiochemotherapy? 3. 57-year-old man with oligometastatic adenocarcimoma of the esophago-gastric junction: uT2 N+ M1 (lymphatic): What are the next steps: Palliative chemotherapy? Curative resection? Multimodal therapy?

108

Oncol Res Treat 2015;38(suppl 5):1–270

4. 75-year old man with metastatic gastric cancer, mixed type according to Lauren, multiple metastases in the liver and bone. What’s next: molecular testing? Chemotherapy first-line? Which choice of first-line treatment? 5. 57-year old man with gastric cancer, diffuse type according to Lauren, treatment with fluorouracil, leucovorin, oxaliplatin, docetaxel (FLOT) first-line for 6 months. Then: progression with massive ascites. What’s next: intraperitoneal therapy? Systemic treatment? Which choice of second-line treatment? I invite you to join this expert seminar in order to solve these difficult as well as interesting cases! Disclosure: Florian Lordick: Employment or Leadership Position: keines; Advisory Role: Ganymed, Biontech, Roche, Taiho, Lilly, Boston Biomedical; Financing of Scientific Research: an die Institution des Autors; Expert Testimony: GSK, Fresenius Biotech; Other Financial Relationships: Reisekostenterstützung für Kongresse: Roche, Bayer, Taiho, Merck.

Fortbildung

NHL aggressiv – Konzepte bei B- und T-Zell Lymphomen V362

Treatment of aggressive B-cell lymphomas: R-CHOP or more? Pfreundschuh M.1, Deutsche Studiengruppe für Hochmaligne  Non-Hodgkin-Lymphome Universität des Saarlandes, Innere Medizin I, Homburg, Germany

1

Young patients with no risk factor according to the age-adjusted (aa) IPI and no bulky disease have been shown in the MInT study to have a high event-free and nearly 100% overall survival after 6 cycles of R-CHOP-21, suggesting that some of these patients are overtreated. For the first time in the history of DLBCL treatment prospective trials like the FLYER study evaluate reduction of treatment from 6 to only 4 cycles of CHOP with 6 applications of rituximab. For young patients with aaIPI = 1 and/ or bulky disease, best results have been achieved with 6xR-CHOP-21 plus involved-field radiotherapy to bulky disease or the more aggressive and more toxic R-ACVBP program without radiotherapy. For young poor-prognosis patients (aaIPI = 2,3) best results have been achieved with 8xR-CHOEP-14, with a 3-year overall survival of aaIPI = 2 patients of roughly 90%, demonstrating that this subpopulation is no high-risk population any more in the rituximab era. For young aaIPI = 3 patients there is still room for improvement and ongoing trials evaluate primary highdose chemotherapy with stem cell transplantation or the addition of new drugs to a CHOP(E)P-14 backbone. Elderly patients who represent up to two thirds of all DLBCL patients do best with 8 cycles of R-CHOP-21 or 6 cycles of R-CHOP-14 plus 2 additional rituximab administrations. Since further intensification of chemotherapy is hardly possible in the elderly population, improvement strategies evaluate new doses and schedules of rituximab based on the recent demonstration that rituximab is suboptimally dosed in all DLBCL patients except elderly females. Other improvement strategies include vitamin D substitution in vitamin D insufficient and deficient DLBCL patients, because vitamin D deficiency impairs ADCC, the most important mechanism of action of rituximab, as well as new drugs targeting specific molecules involved in the signal transduction pathway of the B-cell receptor. There are several novel concepts in clinical trials, the most promising being BTK and BCL2 inhibitors, with BARs (B-cell receptor antigens for reverse targeting) providing an ultimate specificity restricted to the malignant B-cell clone. While the rate of patients dying from DLBCL has been cut into half during the last decade, these new approaches should further improve the cure rate of DLBCL in the near future. Disclosure: Michael Pfreundschuh: Advisory Role: Advisory Board.

Abstracts


CONTENTS AUTHOR INDEX

Wissenschaftliches Symposium CLL I V365

Cellular origin of CLL Küppers R.1 Universität Duisburg-Essen, Institut für Zellbiologie (Tumorforschung), Essen, Germany 1

In chronic lymphocytic leukemia (CLL), about half of the cases carry mutated immunoglobulin V region genes, and the other half are unmutated. Gene expression profiling studies revealed that both groups show a highly similar gene expression pattern. The cellular origin of CLL is much discussed and still not fully resolved. A derivation from CD5-positive B cells had originally been proposed. However, also marginal zone B cells, B cells activated in T independent immune responses and memory B cells are considered as more likely cellular origin of this most frequent leukemia in adults. We performed a refined gene expression profiling study of CLL cells in comparison to naive, mature CD5+, IgM+IgD+CD27+, IgM-only, class-switched memory and splenic marginal zone B cells. The evaluation of the data indicates that the gene expression program of both groups of CLL is most similar to mature CD5+ B cells, suggesting that CLL derive from these cells. We also identified a small subset of CD5+ B cells with somatically mutated immunoglobulin VH genes, which is characterized by expression of the memory B cell marker CD27. The mutated CLL tended to be more similar in their gene expression to these mutated CD5+ B cells than to unmutated CD5+ B cells, suggesting that mutated CLL originate from these germinal center-experienced CD5+ memory B cells. The proposed origin of CLL from mature CD5+ B cells is also supported by the detection of stereotyped immunoglobulin VH gene rearrangements among normal CD5+ B cells. Moreover, our study revealed a distinct gene expression pattern for mature CD5+ B cells, indicating that these lymphocytes represent a distinct B cell differentiation stage, and perhaps have specific functions in immune responses. Disclosure: No conflict of interest disclosed. V367

Activation of autonomous signaling in CLL by mutual BCR interaction Jumaa H.1 Universitätsklinikum Ulm, Institut für Immunologie, Ulm, Germany

1

Introduction: Chronic lymphocytic leukemia (CLL) is a heterogeneous malignancy and is defined as a monoclonal expansion of morphologically mature but functionally incompetent B-lymphocytes with a distinct phenotype. Several mechanisms that mediate the malignant phenotype of CLL cells have been identified and suggest that both cell intrinsic factors as well as signals from the microenvironment contribute to the pathogenesis and prognosis of CLL. For instance, typical CLL-associated genetic alterations, which usually affect genes involved in cell cycle regulation or apoptosis, determine the course of the disease and subsequently more aggressive behavior. Increasing evidence strongly suggests an important role of the B cell receptor (BCR) in CLL etiology and since BCR signaling is usually activated by engagement through cognate antigen, it was assumed that external antigens induce BCR signaling and determine the survival of CLL B cells. We have recently shown that CLL-derived BCRs possess the unique capacity of autonomous signaling, which is mediated by mutual BCR interaction on the same cell. Methods: We have performed crystallographic analyses of CLL-derived BCRs and found that the analyzed receptors undergo a specific and direct interaction with each other. We determined and subsequently mutated the crucial amino acid residues and expressed the resulting constructs in our TKO cellular reconstitution system described previously. Results: We found that the original “wild-type” CLL-derived BCR is capable of inducing autonomous signaling in the reconstitution system for

Abstracts

BCR expression, independent of external antigens. In contrast, however, the derivatives in which the amino acid residues which are crucial for interaction according to crystallography were mutated lost the ability for autonomous signaling. Treatment with anti-BCR antibody induced the signaling of the mutated version. Conclusion: The autonomous signaling capacity is induced by direct interaction of BCR molecules in the membrane of the same cell and is characteristic for CLL. Identification of the crucial residues for this interaction might the design of specific reagent that may interfere with the autonomous signaling capacity and prevent CLL progression. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium Frühes Prostatakarzinom V369

Risk-adapted PSA screening – a way to reduce the harms of prostate cancer screening Recker F.1, Randazzo M.1,2, Wyler S.F.1, Huber A.3, Grobholz R.4, Manka L.5, Kwiatkowski M.1,5 Kantonsspital Aarau AG, Klinik für Urologie, Aarau, Switzerland, Universitätsspital, Zürich, Switzerland, 3Kantonsspital Aarau AG, Zentrum für Labormedizin, Aarau, Switzerland, 4Kantonsspital Aarau AG, Institut für Pathologie, Aarau, Switzerland, 5Akademisches Lehrkrankenhaus, Klinik für Urologie, Braunschweig, Germany 1 2

Introduction: PSA-Screening using a static PSA-cutoff for prostate biopsy (PBx) reduces prostate cancer (PCa)-specific mortality but is associated with a high rate of overdiagnosis of 50%. Treatment of overdiagnosed PCa may reduce men`s quality of life, known as “screening harms“. This in turn underlines the unmet need for a risk-based PSA-screening strategy. The current work shows the development of the Swiss PCa risk calculator ProstateCheck© in order to perform a “step-by-step” risk-adapted PSA-Screening. Methods: From 1998 to 2012, 4932 men were prospectively observed (ERSPC Aarau, Switzerland). PSA-screening was undertaken every 4 years with a value of ≥3.0 ng/ml as indication for PBx. At baseline, men with 1–2.99 ng/ml and free-to-total PSA-Ratio (f/t-PSA) = < 20% also underwent PBx. Primary endpoint was PCa diagnosis. Aggressive PCa was defined as intermediate and high-risk PCa according to d`Amico classification. Hosmer-Lemeshow-Test was used to count for model variables. The area under the curve (AUC) was calculated. The model was divided into a diagnostic (PSA > = 3 ng/ml) as well as a prognostic (PSA < 3 ng/ml) component. Results: The best prediction model was achieved by using the following combined predictors: age, PSA-value, f/t-PSA, family history, prostate volume and digital rectal examination (DRE). The AUC was 0.74 (95% CI 0.69–0.80) for overall PCa diagnosis. For detection of aggressive PCa, AUC increased as follows: 0.72 (PSA), 0.80 (PSA+f/t-PSA), 0.82 (PSA+DRE), 0.87 (PSA+DRE+f/t-PSA), 0.88 (PSA+DRE+f/t-PSA+Age), 0.89 (PSA+DRE+f/t-PSA+age+family history+ prostate volume). Among men with baseline PSA < 3ng/ml, the incidence of aggressive PCa was recorded during follow-up. Using these frequencies, re-test intervals for subsequent PSA-screening according to baseline PSA were calculated for an individualized screening algorithm. Conclusion: The Swiss PCa risk calculator ProstateCheck© can predict the probability of aggressive PCa with a high AUC (0.89) as compared to other risk tools (AUC of PCPT- /San Antonio 0.63, AUC Rotterdam 0.82). Aside from its diagnostic abilities, an individualized re-screening interval can be proposed which can save up to 70% of unnecessary PSA-tests and therefore reduce overdiagnosis. With this refinement, both screening and decision to perform PBx can be adjusted according to a man`s individual risk constellation leading to a more personalized medicine.

Oncol Res Treat 2015;38(suppl 5):1–270

109


CONTENTS AUTHOR INDEX

Disclosure: Franz Recker: No conflict of interest disclosed. Maciej Kwiatkowski: Advisory Role: Astellas und Myriad. V371

The PREFERE trial – the desire to prove the efficacy of treatment options for low risk prostate carcinoma Weissbach L.1 Stiftung Männergesundheit, Berlin, Germany

1

The German S3 guideline recommends in treating low risk carcinoma of the prostate: prostatectomy, irradiation, LDR brachytherapy, active surveillance. Considering these recommendations insufficiently substantiated, the initiators of PREFERE started the trial stating that ‘the recommendations of the S3 guideline appear to suggest that the four alternative treatments are equally effective in tumour control. This is definitively not the case. There are no conclusive data proving this assumption’ (Michael Stöckle, principal investigator). However, the epidemiology of low risk prostate carcinoma as well as results of already published or other initiated RCTs comparing treatment options of localized prostate carcinoma may raise doubts if the clinical superiority of one or the other option can, or even need to be proven. The mortality of low risk PCa (2% within 15 years subsequential early detection of the tumor) is very low, and reliable data concerning the mortality of prognostically particularly favourable low volume tumours are still lacking. Even a study population of 7,600 men is thus unlikely to yield statistically significant differences with PCa mortality as primary endpoint. In addition, results of PIVOT (though methodically not entirely flawless) show no significant advantage of radical prostatectomy as compared to no treatment. As PREFERE sets out to include only men with PSA ≤10 ng/ml the hope to find a survival benefit with one of the treatment modalities in comparison to any other of the available options seems highly questionable. Experiences of already initiated studies indicate considerable recruitment difficulties: PIVOT recruited only 700 men instead of 5,000 planed for, START had to be terminated for lack of sufficient numbers, and PROTECT succeeded in recruiting only half of the anticipated study population. Obviously, available information about benefits and risks of available treatment options are sufficient to prompt potential candidates to refuse randomisation. PREFERE’s problems with recruiting patients do not imply a general impossibility to conduct RCTs nor do they exclude the potential of other study designs to answer questions of efficacy. They do however indicate the fact that potential patients as well as some treating urologists may have different views than the initiators of PREFERE concerning the need of comparative therapy studies of low risk PCa – an experience common to this and other large trials investigating its treatment Disclosure: No conflict of interest disclosed.

Fortbildung

Geriatrische Onkologie Hämatologische Erkrankungen im Alter 80+ V377

Elderly patients with diffuse large B-cell-lymphoma Zettl F.1, Hohloch K.2, Trümper L.3 Klinikum Traunstein, Hämatologie/Onkologie und Palliativmedizin, Traunstein, Germany, 2Department Hämatologie und Onkologie, Kantonsspital, Chur, Switzerland, 3Klinik für Hämatologie und Medizinische Onkologie und Stammzelltransplantation, Universitätsmedizin, Göttingen, Germany 1

Introduction: Aggressive Lymphoma is a frequent disease in elderly patients, the median age at diagnosis is 66 years. Therapeutic decisions for elderly patients are difficult to make and often influenced by other factors like social status and physicians’ experience and preferences. Raising

110

Oncol Res Treat 2015;38(suppl 5):1–270

numbers of comorbidities are complicating the treatment and management of elderly patients with aggressive lymphoma. Results/Discussion: Poor risk factors identified in a SEER Database analysis in elderly patients are age above 80 years, pre-existing heartfailure and an increased comorbidity score. Nevertheless in elderly and comorbid patients the intention of treatment must be curative, the only curative treatment approach with a 5-year overall survival of approx. 75% is a combined immuno-chemotherapy including anthracyclines. The cumulative amount of antracylines given directly correlates with prognosis; however comorbidities are often the limiting factor of a full dosed CHOP chemotherapy. A dose reduced CHOP (mini-CHOP) in combination with rituximab has shown acceptable efficacy with reduced toxicity in patients not qualifying for fulldose therapy. The decision if such a regimen is feasible can only be made after a prephase therapy in individual patients. In addition, comorbidities and socioeconomic factors must be incorporated into the treatment plan in this patient population. Geriatric assessments may aid in the establishment of rational approaches. In common practice, Bendamustine is frequently being used in frail patients not qualifying for an antracyline based therapy. There is limited prospective data concerning the efficacy of this approach. Studies are ongoing. Conclusion: Therapeutic options in elderly patients are very limited. Antracyclines are essential for a curative treatment strategy. Probably standardized geriatric assessments can help identify patients who are feasible for a more aggressive approach. The final choice of treatment has to be made by the treating physician in close interaction with the patient and his relatives. Disclosure: Florian Zettl: Expert Testimony: Drittmittelföderung durch Roche und Mundipharma. Lorenz Trümper: Expert Testimony: Drittmittelföderung durch Roche und ­Mundipharma. V378

Patients with Multiple myeloma Kleber M.1 Clinic for Internal Medicine, University Hospital, Basel, Switzerland

1

Multiple myeloma (MM) is a disease which primarily affects elderly patients with a median age of 70 years. Over the last decade, novel therapeutic agents and enhanced supportive care improved survival in MM. Though, this benefit in survival appears to be confined to the young: the 5-year relative survival for MM patients younger than 65 years improved by more than 17% between 1998 and 2002 and between 2003 and 2007, but only 3.3% in the elderly (≥75 years). To date, the choice of therapy in MM patients is primarily based on chronologic age and performance status (PS). However, recent studies in MM highlight that the biological fitness of elderly patients shows relevant variations and assessment strategies are essential to define functional status, comorbidities and vulnerability to adverse events, which may increase treatment toxicity and drug discontinuation. A number of prognostic scores for evaluating comorbidity in cancer patients are available. Especially in MM specific comorbidity scores are proposed. E.g. the Freiburg Comorbidity Index (incl.: Karnofsky PS, renal and pulmonary disease status) is a reliable tool and highly predictive for outcome. Nevertheless, a systematic and evidence-based way of defining the global health and functional status is desirable to guide and objectify treatment decisions. In cancer patients a Comprehensive Geriatric Assessment (CGA) is considered to be the most appropriate way to evaluate the functional status, frailty and cognitive impairment. In MM patients the CGA is not routinely performed. Recently, Italian authors performed a brief geriatric assessment (GA) in 869 elderly patients with newly diagnosed MM. The GA consisted of 3 tools (ADL, IADL and Charlson Comorbidity Index). The results showed that a GA combined in a frailty score including age, comorbidities, cognitive and physical conditions can identify 3 groups of fit, intermediate fitness and frail patients with different 3-year OS of 84%, 76% and 57%, regardless of staging and treatment administered. In addition, frailty was associated with an increased risk of nonhematologic AEs and treatment discontinuation. Further studies

Abstracts


CONTENTS AUTHOR INDEX

in MM to validate determinants of therapeutic decision-making, in elderly-specific MM therapeutic and interventional trials with relevant outcomes beyond survival (e.g. effects of long-term toxicity on quality of life, independence, etc.) are needed. Algorithms for optimal therapy allocation in older patients will then be warranted. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium Stem Cell Biology – New Insights

Freier Vortrag

Kolon-/Rektumkarzinom I V390

Next-generation sequencing liquid biopsy strategy to monitor the emergence of EGFR antibody-resistant tumor subclones in gastrointestinal cancer Braig F.1, März M.1, Schieferdecker A.1, Schulte A.2, Voigt M.1, Stein A.1, Grob T.3, Alawi M.4, Indenbirken D.5, Kriegs M.6, Engel E.7, Vanhoefer U.8, Grundhoff A.5, Loges S.1,9, Riecken K.10, Fehse B.10, Bokemeyer C.1, Binder M.1 Universitätsklinikum Hamburg-Eppendorf, Onkologisches Zentrum, II. Medizinische Klinik und Poliklinik, Hamburg, Germany, 2Universitätsklinikum Hamburg-Eppendorf, Kopf- und Neurozentrum, Klinik und Poliklinik für Neurochirurgie, Hamburg, Germany, 3Universitätsklinikum HamburgEppendorf, Zentrum für Diagnostik, Institut für Pathologie, Hamburg, Germany, 4Universitätsklinikum Hamburg-Eppendorf, Bioinformatics Service Facility (BIS), Hamburg, Germany, 5Heinrich-Pette-Institut, Leibnitz-Institut für experimentelle Virologie, Hamburg, Germany, 6Universitätsklinikum HamburgEppendorf, Onkologisches Zentrum, Klinik und Poliklinik für Strahlentherapie und Radioonkologie, Hamburg, Germany, 7Hämatologisch-onkologische Praxis Altona (HOPA), Hamburg, Germany, 8Marienkrankenhaus, Zentrum für Innere Medizin, Hamburg, Germany, 9Universitätsklinikum Hamburg-Eppendorf, Zentrum für Experimentelle Medizin, Institut für Tumorbiologie, Hamburg, Germany, 10Universitätsklinikum Hamburg-Eppendorf, Onkologisches Zentrum, Klinik für Stammzelltransplantation (KMT), Hamburg, Germany 1

V385

Mathematical modelling of benign and malignant hematopoiesis: Potentials and insights Roeder I.1 Medizinische Fakultät Carl Gustav Carus an der Technischen Universität Dresden, Institut für Medizinische Informatik und Biometrie, Dresden, Germany 1

Systems biology, i.e. the application of mathematical models to quantitatively describe complex biological systems, is still a nascent discipline. However, even in the context of translational and clinical applications, the use of computational methods is becoming more and more accepted. In my presentation I will discuss the potentials of mathematical modeling and simulation studies to broaden our understanding of hematopoietic stem cell organization. This comprises processes on the cellular level, such as cell proliferation or migration within the so called bone marrow stem cell niche, the tissue level, such as clonal competition, and last but not least, on the patient level, such as the prediction of treatment effects in chronic myeloid leukemia. Using selected examples of modeling results, I will demonstrate how theoretical approaches can help to disentangle competing hypotheses about biological processes and to support the design of optimal experimental and/or clinical strategies. On the other hand, I will also point to potential pitfalls and limitations of in silico studies. Disclosure: Ingo Roeder: Financing of Scientific Research: Bristol-Myers Squibb; Expert Testimony: Bristol-Myers Squibb. V386

Designer nuclease mediated gene modulation in stem cells Cathomen T.1 Universitätsklinikum Freiburg, Institut für Zell- und Gentherapie, Freiburg, Germany 1

Targeted genome editing with designer nucleases has become increasingly popular. The most commonly used designer nuclease platforms are meganucleases, zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas system. These powerful tools have greatly facilitated the generation of plant and animal models for basic research, and harbor an enormous potential for applications in biotechnology and regenerative medicine. However, the application of designer nuclease technology in human stem cells requires special considerations. Given that off-target cleavage activity of customized nucleases can potentially induce genotoxic side-effects, including the transformation of the target cell type, particular attention has to be paid to the specificity of designer nucleases. In my talk I will present data on preclinical applications of designer nucleases, highlight parameters that affect specificity of these nucleases, and end with a personal view on what aspects should be contemplated before moving into the clinic.

Introduction: The emergence of epidermal growth factor receptor (EGFR) ectodomain mutations as well as activating RAS mutations represents a clinical challenge in EGFR targeting of colon cancer. However, screening for such mutant subclones hasn’t entered routine clinical practice yet. Methods: 21 solid tumor samples of patients suffering from gastrointestinal cancer treated with chemotherapy +/- EGFR antibodies cetuximab or panitumumab were screened by next generation sequencing (NGS) for mutations in KRAS exon 2/3/4, NRAS exon 2/3/4 and the overlapping epitopes of both EGFR antibodies. Results were validated using circulating tumor DNA (ctDNA) of an independent cohort of 27 patients. Binding, signaling and drug sensitivity studies were performed in Ba/F3 cells to functionally characterize the EGFR G465R mutation. Results: In these two patient cohorts that were in their majority still responsive to the combination of chemotherapy with EGFR antibodies, we found the EGFR G465R mutation in 7% and activating RAS mutations in 21% of cases. In line with previous data, this suggests that these mutations may be picked up even before overt clinical resistance occurs. NGS screening of ctDNA seemed to be more sensitive than NGS of post-treatment tumor material in the detection of such mutations, most likely because ctDNA more broadly reflects tumor heterogeneity. The EGFR G465R mutation was characterized for loss of cetuximab and panitumumab binding and for induction of cross-resistance in EGFR G465R transfected Ba/F3 cells. Conclusions: We show that EGFR ectodomain mutations may also emerge in patients with gastrointestinal cancers other than colorectal cancer (e.g. cholangiocellular carcinoma) under the selective pressure of EGFR antibodies. These mutations can be easily screened for and monitored by a next-generation sequencing liquid biopsy strategy using ctDNA. This technology may compare favorably to BEAMing in terms of cost-effectiveness. Prospective studies should address what minimal mutational loads are required to faithfully predict lack of response to EGFR antibodies. Disclosure: No conflict of interest disclosed.

Disclosure: Toni Cathomen: Advisory Role: Beratungstätigkeit für TRACR Hematology Ltd.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

111


CONTENTS AUTHOR INDEX

V391

V392

Molecular profiling of single circulating colon cancer cells (liquid biopsy)

AIO-KRK-0109: A randomized phase II trial of panitumumab plus FOLFOXIRI or FOLFOXIRI alone as 1st-line treatment in RAS-wild-type metastatic colorectal cancer (mCRC)

Liebs S.1, Keilholz U.2, OncoTrack Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Berlin, Berlin, Germany, 2 Charite Universitätsmedizin Berlin, Berlin, Germany

Martens U.1, Weßendorf S.2, Riera Knorrenschild J.3, Büchner-Steudel P.4, Florschütz A.5, Atzpodien J.6, Keller R.7, Greeve J.8, Kanzler S.9, Ettrich T.10, Lindig U.11, Egger M.12, Hebart H.13, Geißler M.2, AIO

Characterization of circulating tumor cells (CTCs) from tumor patient`s blood (liquid biopsy) represents a non-invasive approach for cancer profiling and treatment monitoring. We established a protocol for single CTC isolation based on the distinction of CD45-positive leukocytes and tumor cells expressing the epithelial markers EpCAM and/or carcinoembryonic antigen (CEA). The method is successfully implemented into the EU OncoTrack Project designed to identify novel biomarkers for colon cancer. Prior to surgery, 50–60 ml of peripheral blood from patients with primary or metastatic colon cancer was collected. After red blood cell lysis, negative enrichment of CTCs was performed using the slightly modified EasySep(TM) human CD45 Depletion Kit protocol (StemCell). Immunofluorescent staining was used to detect and aspirate viable (LIVE/ DEAD-negative), EpCAM/CEA-positive and CD45-negative CTCs by microscopic-assisted micromanipulation. Linear RNA amplification of the cell lysate and subsequent RT-qPCR was performed using CD45 and cytokeratin 19 (CK19), an intracellular tumor marker that is not downregulated during epithelial-to-mesenchymal transition. Application of the protocol allowed CTC detection in 21 of 39 (54%) blood samples of patients with primary colon cancer and in 15 of 25 (60%) with metastases. A CTC detection rate of 88% was achieved in blood samples from patients undergoing surgery of the primary and metastatic tumor. The number of isolated cells varied between 1–4 cells (primary tumor) and 1–7 CTCs (metastatic disease) per blood sample. TaqMan and HybProbe PCR showed inconsistent results probably due to more successful amplification of smaller target regions on fragmented sample cDNA (e.g. CK19 amplicon length of 90 bp and 150 bp, respectively). Based on their positive CK19 and housekeeping gene status, 10 of 32 (31%) TaqMan or HybProbe-analyzed cells were defined as CTCs. Only 1 of 26 TaqMan-analyzed cells showed CD45 expression proving the accuracy of the cell picking. A panel deep sequencing protocol for targeted mutational analysis is under development. CTC analysis holds tremendous potential for the identification of new drug targets and resistance-associated markers. Therefore, the introduced method represents a promising approach, which is not only adaptable to other tumor entities but also allows a variety of downstream applications.

1

1

Disclosure: No conflict of interest disclosed.

SLK-Kliniken Heilbronn GmbH, Medizinische Klinik III, Heilbronn, Germany, Klinikum Esslingen GmbH, Klinik für Allgemeine Innere Medizin, Onkologie/ Hämatologie, Gastroenterologie und Infektiologie, Esslingen, Germany, 3 Universitätsklinikum Gießen und Marburg GmbH, Hämatologie, Onkologie und Immunologie, Marburg, Germany, 4Universitätsklinikum Halle (Saale), Uniklinik und Poliklinik für Innere Meidzin I, Halle, Germany, 5Städtisches Klinikum Dessau, Klinik für Innere Medizin, Dessau-Roßlau, Germany, 6 Franziskus Hospital Nils-Stensen-Kliniken, Klinik für Interne Onkologie und Hämatologie, Georgsmarienhütte, Germany, 7AIO-Studien-gGmbH, Berlin, Germany, 8St. Vincenz Krankenhaus Paderborn, Medizinische Klinik I und Vincenz Darmzentrum, Paderborn, Germany, 9Leopoldina-Krankenhaus d. Stadt Schweinfurt GmbH, Medizinische Klinik II, Schweinfurt, Germany, 10 Universitätsklinikum Ulm, Klinik für Innere Medizin I, Ulm, Germany, 11 Universitätsklinikum Jena / Institut für Psychosoziale Medizin und Psychotherapie, Klinik für Innere Medizin I, Jena, Germany, 12Ortenau Klinikum, Hämatologie / Onkologie, Lahr, Germany, 13Stauferklinikum Schwäbisch Gmünd, Klinik für Innere Medizin, Mutlangen, Germany 2

Background: Patients with mCRC belonging to ESMO groups 1 and 2 are recommended to be treated with the most active applicable chemotherapy aiming on rapid and effective tumor responses. Group 1 patients may have the chance of a curative secondary resection of liver/lung metastases while group 2 patients have tumor symptoms and/or the presence of an aggressive tumor biology. The current study evaluates the efficacy of FOLFOXIRI plus panitumumab compared to FOLFOXIRI alone. Methods: Prospective open-label 2:1 randomized Fleming’s single stage phase II study using Irinotecan 150 mg/m2 d1; Oxaliplatin 85 mg/m2 d1; Leucovorin 200 mg/m2 d1;5-FU 3000 mg/m2 cont. 48 h; Panitumumab 6.0 mg/kg q2w (arm A) and Irinotecan 165 mg/m2 d1; Oxaliplatin 85 mg/ m2 d1; LV 200 mg/m2 d1; 5-FU 3200 mg/m2 cont. 48 h q2w (arm B) up to 12 cycles. Primary endpoint was objective RR; secondary endpoints were secondary tumor resection rate, DCR, PFS, TTR, OS, toxicity, QL. To date 72/93 patients are recruited. Results: A planned interim analysis was performed after 30 patients treated in Arm A to demonstrate a CR/PR in >18 of treated patients. 10 (33.3%) and 20 (66.7%) patients belonged to ESMO groups 1 and 2, respectively. Median age was 55.5 years (31–76 years). A median of 8.5 cycles FOLFOXIRI+Panitumumab were applied. The objective RR was 86.7% (n = 26). DCR was achieved in 30 patients (100%). A preplanned safety analyses after 10 patients in arm A did result in a dose adaption of irinotecan from 165 to 150 mg/m2 and 5-FU from 3200 mg to 3000 mg/ m2. With this dosage no severe safety signals were observed compared to FOLFOXIRI alone although there was an increased number of SAE events.. Discussion: After dose modification of the FOLFOXIRI regimen the addition of panitumumab had a tolerable safety profile. A strong efficacy signal regarding RR and DCR was observed suggesting that FOLFOXIRI+panitumumab may be a prefered treatment for selected RAS wild-type patients belonging to ESMO groups 1+2. Further data will be presented. Disclosure: No conflict of interest disclosed.

112

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V393

The colorectal carcinoma – treatment research and treatment reality in oncology practices

Tab. 3. response

Valdix A.-R.1, Zaun S.2, Göttel R.3, Tessen H.-W.4,5 Onkologische Schwerpunktpraxis, Schwerin, Germany, Sanofi-Aventis Deutschland GmbH, Berlin, Germany, 3rgb Onkologisches Management GmbH, Sarstedt, Germany, 4Onkologische Kooperation Harz, Goslar, Germany, 5 Projektgruppe Internistische Onkologie (PIO), Goslar, Germany 1

2

Introduction: Which contribution does a continuous, systematic case documentation and evaluation provide to treatment research? What is the significance of new substances such as Aflibercept? Methods: Data from oncology practices from 2003–2015 (PIO, ONCOReg). 7,802 histories of diseases comprising 12,623 palliative treatments originating from 123 oncology practices from 16 federal states are available for analysis. 12,446 treatments have been concluded so far and a response has been recorded in 11,655 of them. Results: Patient characteristics Gender: m: 60%, f: 40% Distant metastatic diseases: 5,151 patients (85% lung/liver) KRAS mutation status (n = 1,957): 63% wild type, 37% mutant Median age at start of 1st-line therapy: 68 (19–92) years Palliative chemotherapy (n = 5,076): Tab. 1. Therapy

%

1st-line

100

2nd-line

68

3rd-line

37

4th-line

18

OR

NC

PD

n.k.

overall (n = 67)

10

28

39

22

1st-line (n = 3)

33

33

0

33

2nd-line (n = 21)

9

38

48

5

3rd-line (n = 10)

20

20

40

20

4th-line (n = 17)

13

29

29

29

Conclusions: The data collected over a long period of time depict very precisely the reality of treatment in oncology practices in Germany. The analysis presented provides an important contribution to the complex of treatment research and answers a series of patient-related questions. Aflibercept is applied according to its approved use. Current data will be presented. Disclosure: No conflict of interest disclosed. V394

Implementation of RAS testing in patients with metastatic colorectal cancer in German outpatient cancer centers – data from the clinical TKK registry Schnell R.1, Trarbach T.2, Broszeit-Luft S.3, Musch R.4, Frank M.2, Kopfmann S.2, Jänicke M.2, Marschner N.5, for the TKK Registry Group Praxis Internistischer Onkologie und Hämatologie, Frechen, Germany, iOMEDICO, Freiburg, Germany, 3Onkologische Praxis, Lehrte, Germany, 4 Krebsheilkunde Lichtenberg, Berlin, Germany, 5Praxis für Interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany 1

Median duration of treatment (n = 11,446): 119 days

2

Tab. 2. Response (n = 11,655)

%

OR

NC

PD

n.k.

overall

28

38

23

11

1st-line

41

37

12

10

2nd-line

23

41

26

10

3rd-line

16

37

34

13

4th-line

11

35

40

14

Survival PFS/OS as of start of 1st-line treatment: 9.5/24.7 mths. PFS/OS as of start of 2nd-line treatment: 6.4/15.1 mths. 27 practices treated 98 patients with aflibercept (87% with FOLFIRI). Median age at start of 1st-line treatment: 74 years. 32% of patients received 2nd-line, 22% 3rd-line, 20% 4th-line. 97% of patients had been pretreated with oxaliplatin. Median duration of aflibercept-based therapy (n = 65): 73 days

Abstracts

%

Introduction: Since 2008 KRAS mutation testing is mandatory before applying an anti-EGFR monoclonal antibody (mab) in patients (pts) with metastatic colorectal cancer (mCRC). Since 2014 expanded RAS mutation testing including exons 2–4 of the KRAS gene as well as exons 2–4 of the NRAS gene is required. Methods: Starting in 2006, the Tumor Registry Colorectal Cancer (TKK) prospectively documents treatment of pts with CRC by office-based medical oncologists in Germany. Pts receive standard treatments according to physician’s choice. The registry collects data on biomarker testing, all systemic treatments, outcome, treatment decision-making as well as pts and tumor characteristics. Currently (March 2015), 166 sites have recruited over 5500 pts with CRC, of which about 3700 have mCRC. Information on mutation testing has been collected for KRAS status since 2008 (n = 2264) and for RAS since 2014 (n = 226). Results: The number of pts tested for KRAS mutation before the start of first-line treatment has been increasing since 2008 (47% in 2008, 86% in 2015). NRAS status was determined in 58% of pts in 2014, and in 63% in early 2015. These results correlate with the increasing number of pts tested for both RAS mutations (58% in 2014, 62% in 2015). Pts who are not tested for RAS mutation are older (70 vs. 67 years) and more often affected by a concomitant disease (83% vs. 72%) compared to pts tested for RAS mutation. The main reason for not testing pts’ RAS status before the start of first-line treatment is “no intention to use an anti-EGFR mab”, according to the attending oncologists. While 54% of pts tested were KRAS wild-type (wt) in 2014, this number slightly decreased to 48% in 2015, presumably because of more exons are being tested. This trend is also seen in pts tested for NRAS mutation (89% (n = 156) wt in 2014 and 83% (n = 46) in 2015). Of the 226 pts tested for

Oncol Res Treat 2015;38(suppl 5):1–270

113


CONTENTS AUTHOR INDEX

both, NRAS and KRAS mutation, 69% were RAS wt in 2014 and 57% in 2015. Of all pts with RAS wt status, 51% were subsequently treated with anti-EGFR mab-based regimen as first-line treatment, most frequently with FOLFIRI and cetuximab (38%). The 49% of pts not treated with an anti-EGFR mab, most frequently received FOLFIRI and bevacizumab (28% of RAS wt pts). Conclusion: Testing for RAS mutation has been rapidly implemented in German outpatient cancer centers with almost all pts being tested for KRAS and an increasing number of pts being tested for NRAS. Disclosure: No conflict of interest disclosed. V395

Relevance of neo- or adjuvant pretreatment in patients with metastatic colorectal cancer treated with FOLFIRI plus bevacizumab as first-line treatment – data from the prospective TKK registry Schnell R.1, Trarbach T.2, Broszeit-Luft S.3, Musch R.4, Wetzel N.2, Kopfmann S.2, Jänicke M.2, Marschner N.5, for the TKK Registry Group Praxis Internistischer Onkologie und Hämatologie, Frechen, Germany, iOMEDICO, Freiburg, Germany, 3Onkologische Praxis, Lehrte, Germany, 4 Krebsheilkunde Lichtenberg, Berlin, Germany, 5Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany 1 2

Background: A large number of patients (pts) diagnosed with metastatic colorectal cancer (mCRC) have received neo- or adjuvant treatment for initially localized CRC. The influence of such pretreatment on the outcome after diagnosis of metastasis is still under debate. While clinical trials recruit highly selected pts, clinical registries collect data on treatment in routine practice, representing all pts and therapies. This allows analysis of pretreatments and their impact on outcome in real-life pts. Methods: Starting in 2006, the Tumor Registry Colorectal Cancer (TKK) prospectively documents treatment of pts with CRC by office-based medical oncologists in Germany. Pts receive standard treatments according to physician’s choice. A broad set of data including pts and tumor characteristics, all systemic treatments and outcome are collected. At the time of this interim analysis (March 2014), 122 sites had recruited 5024 pts with CRC; 2466 pts received treatment for mCRC. Here, outcome data such as progression free survival (PFS) and overall survival (OS) of pts treated with the most frequently applied first-line treatment FOLFIRI and bevacizumab (FOLFIRI+BEV, 24% [n = 588]) were estimated. Progression was reported by study sites as per local standard. The outcome was compared between pts pretreated with neo- or adjuvant chemotherapy (pretreated pts) and treatment naïve pts (naïve pts). Results: At the start of first-line treatment, 36% (n = 211) of pts receiving FOLFIRI+BEV were pretreated (64% naïve). Pts characteristic were comparable between pretreated and naïve pts: 65% of pts were male (vs. 68%), mean age was 64 years (vs. 64 years), 71% of pts had a concomitant disease (vs. 62%). According to the attending physician, 50% of pretreated pts had a complete or partial response (vs. 56%). Median first-line PFS of pretreated pts was 10.9 months (95% confidence interval [CI] 10.1–11.6) and median OS was 24.7 months (95% CI 19.8–29.7). Without neo- or adjuvant pretreatment, median first-line PFS was 10.8 months (95% CI 9.8–11.7) and median OS was 23.4 months (95% CI 21.0–25.8). Conclusions: In routine practice, median OS and PFS of mCRC pts treated first-line with FOLFIRI+BEV do not differ between pts pretreated with neo- or adjuvant chemotherapy and pts who were not. Median OS and PFS of FOLFIRI+BEV are comparable to data from clinical trials although pts are older and more often affected by comorbidities. Disclosure: Roland Schnell: No conflict of interest disclosed. Norbert Marschner: Financing of Scientific Research: Vortragshonorare von Bayer AG, Merck Pharma GmbH, Roche Pharma AG.

114

Oncol Res Treat 2015;38(suppl 5):1–270

Fortbildung

Transplantation Spendersuche GvHD Nachsorge V399

Acute and chronic GVHD: State of the art therapy Greinix H.T.1 Medizinische Universität Graz, Hämatologie, Graz, Austria

1

GVHD has remained a serious and life-threatening complication of allogeneic hematopoietic cell transplantation (HCT).Calcineurin inhibitors (CNI) and methotrexate (MTX) or mycophenolate mofetil (MMF) have been the established GVHD prophylaxis for many years. Sirolimus and CNI demonstrated equal protection from GVHD with less mucosal side effects. Use of antithymocyte globulin in combination with CNI and MTX resulted in significantly reduced incidences of acute GVHD grades III-IV and chronic GVHD with no difference in survival. Recently, post-transplant cyclophosphamide was successfully investigated not only after haploidentical but also HLA-identical related and unrelated donor HCT. Established first-line therapy of acute GVHD consists of corticosteroids at 2 mg/kg/day and no advantages of higher steroid doses have been reported. However, in acute GVHD grade IIa steroid doses of 0.5 mg/kg/day were effective whereas in grades ≥IIb doses of 1 mg/kg/day increased the need for secondary immunosuppressive treatment and thus, are not recommended. Response to first-line therapy assessed on day 14 is significantly correlated to NRM and survival. Second-line therapy of acute steroid-refractory GVHD is indicated after 3 days with progression, 1 week with persistent unimproving grade III GVHD or after 2 weeks with persistent unimproving grade II GVHD. Currently, no standard salvage treatment options for steroid-refractory acute GVHD patients are available and choice of therapy should be guided by potential toxicity, infections and interactions with other agents. Due to its efficacy and excellent safety profile ECP is a valuable second-line treatment option. Patients with NIH-defined moderate or severe chronic GVHD should receive systemic immunosuppressive treatment whereas in mild chronic GVHD topical therapy is preferable. Established first-line treatment of chronic GVHD consists of corticosteroids with or without CNIs. In case of progression under 1 mg/kg/day of steroids for 2 weeks or stable disease on ≥0.5 mg/ kg/day for 4 to 8 weeks salvage therapy should be considered. ECP is recommended for second-line treatment based on efficacy in prospective studies and lack of general immunosuppression. Other salvage therapies include mTOR-inhibitors, MMF, imatinib and rituximab.Supportive care including prophylaxis of infections and osteoporosis, vaccinations, psychosocial and dietary support and rehabilitation are of major importance for patients’ outcome. Disclosure: Hildegard Greinix: Financing of Scientific Research: Therakos.

Fortbildung

AYA Adoleszente und junge Erwachsene – was ist anders? V400

What are the psychosocial challenges after cancer acute treatment for adolescent and young adults? Leuteritz K.1 Universitätsklinikum Leipzig AöR, Department für Psychische Gesundheit, Abteilung für Medizinische Psychologie und Medizinische Soziologie, Leipzig, Germany 1

Introduction: In Germany, about 3% of all cancer diagnoses per year are among adolescent and young adults (AYA). AYAs are dealing with a serious disease at a complex psychosocial stage in life, including development tasks like detachment from the parental home, establishing financial and social independence, the formation and consolidation of a partnership and family as well as starting a professional career. Workingship addresses

Abstracts


CONTENTS AUTHOR INDEX

many aspects of cancer patients´ life satisfaction. The psychosocial impact of cancer in this age group is rarely represented. Methods: A total of N = 354 diagnosed young adult cancer patients who were aged 18–39 years at the time of survey and had completed their acute treatment (range 0 to 6 month) answered a questionnaire, including standardized instruments and self-developed items focusing on life satisfaction, the supply situation and the need for support in addition to medical, sociodemographical and work specific variables. Life satisfaction was measured with the questionnaire for life satisfaction (FLZ) who captures 10 relevant aspects of life. For analysis we considered patients who were “at work/school” before diagnosis (N = 190) and not unemployed/ disabled. Results: Over half of the responding AYAs working/in school before diagnosis (N = 190, age: 28,8 years, men: 29,6%) are satisfied with their life situation (51,7%). They experience most changes after cancer acute treatment in areas of healthiness (61%) followed by changes in terms of leisure activities (49%), professional life (48%) and family planning (46%) as well as financial situation (42%). Dissatisfaction among AYA patients especially concerns areas of family planning (43%), financial situation (42%), sexuality (35%) and professional life (33%). Most satisfaction exists regarding apartment situation, relatives, friends and partnership. Higher life satisfaction is associated with not having operation, working part time before diagnosis, less changes in income since diagnosis as well as low psychological distress and having social support. Sex, diagnosis, number of medical therapies had no significant impact (R²= 0,79). Conclusions: Despite a high life satisfaction after cancer acute treatment more than every third AYA patient reports about dissatisfaction in issues like family planning, financial situation, sexuality and professional life. A future task must be to develop specific interventions in these areas for this age group and their specific problems in Germany. Disclosure: No conflict of interest disclosed. V402

Back to normal life – should a specialized rehabilitation be offered? König V.1 Klinik Bad Oexen, Bad Oeynhausen, Germany

1

Intensified treatment strategies are used in cancer therapy of adolescents and young adults (AYA) to improve treatment results. Therefore, functional impairments and therapy-related disorders are often more severe than those observed in older cancer patients. Cancer incidence rates are lower in AYA, therefore young adults are underrepresented in acute care hospitals compared to older adults. In AYA, psychological distress is higher after cancer therapy as professional training and / or partnership planning are incomplete. These factors should be taken into account in aftercare planning. In-patient rehabilitation in a group setting promises several advantages: Physical recovery is enhanced by the motivational effects among the group members competing against one another. Under professional supervision, the direct exchange among peers is promoted to the advantage that the psychological group meetings have co-therapeutic effects. Additionally, one-on-one consultations are available for topics that have to be dealt with individually, for example therapy-related infertility. Moreover, reintegration into work life, necessary change of occupation with additional career guidance etc. can be addressed specifically in this setting. Thus, age-specific group rehabilitation within the framework of a specialist clinic is an additional therapeutic tool in the aftercare treatment of juvenile cancer patients providing accelerated recovery from cancer. Disclosure: No conflict of interest disclosed.

Abstracts

Fortbildung

Ösophaguskarzinom Therapiemodalitäten State of the Art V403

State of the art – Treatment of localized oesophageal carcinoma according to tumor stage Stahl M.1 Kliniken Essen-Mitte, Internistische Onkologie und Hämatologie, Essen, Germany 1

Introduction: Patients with carcinomas of the oesophagus and oesophago-gastric junction (OGJ) presenting with localized disease are potentially curable. But cure rates and intensity of therapy are different and strictly dependent on tumour extension. Statements: (1) Carcinomas limited to the mucosa (T1): In tumours without risk factors (signet ring carcinoma, G3, ulceration) complete endoscopic resection is the therapy of choice with high cure rates in mucosal (m1-m3) cancers and also superficially submucosal (sm1 < 500müm) adenocarcinomas (G1/2, <20 mm, L0, V0). (2) Localized early cancer (T1sm2): sm2–3 carcinomas have a risk of lymphatic spread in up to 50%. This is even higher in tumours of T2-category. These tumours should therefore be treated with surgical resection. However, recurrences will occur in up to 70% after complete resection if lymph nodes proved to be involved (pN1–3). Preoperative chemoradiotherapy (CRT) showed to significantly improve local tumour control but not overall survival. However, this procedure is limited by the operative risks particularly in patients with squamous cell cancer (SCC), where relevant numbers of the patients may not tolerate transthoracic oesophagectomy. In these patients definitive chemoradiotherapy can be a treatment option with curative intent. Even more, if the primary tumour is located in the cervical or upper intrathoracic part of the oesophagus. (3) Locally advanced cancer (T3–4): Multimodality approaches are reflecting the state of the art in this clinical situation with surgery proved superiority to other approaches regarding local tumour control and survival. But operative mortality of about 10% must be realized in SCC patients. Therefore, definitive chemoradiation showed similar survival in randomized trials, although local tumour control was hampered by omitting surgery. It remains a matter of debate in adenocarcinomas of the OGJ whether and when preoperative chemoradiotherapy should be preferred to perioperative chemotherapy. Adjuvant therapy appears to be restricted to patients with understaged tumours of pT3– 4pN1–3 category where primary surgery has been performed erroneously. Disclosure: No conflict of interest disclosed. V404

Induction chemotherapy – pro arguments Ruhstaller T.1 Onkologie, St. Gallen, Switzerland

1

Induction chemotherapy is defined as two cycles of chemotherapy before the neoadjuvant or definitive chemoradiation. The specific question of an induction chemotherapy before CRT in oesophageal cancer has hardly been investigated in clinical studies so far, however, this approach has been used in a lot of studies with other focus and it is daily practice in a lot of centres. There are several arguments which militate in favour of induction chemotherapy. The treatment can usually be started suddenly after the diagnosis ( no waiting time for planning the radiotherapy). Most patients are presenting by diagnosis with relevant dysphagia. A direct start with radiotherapy is usually worsening the dysphagia and makes often feeding tubes necessary. With induction chemotherapy the dysphagia is often very fast improving in such a way, that patients are able to eat quite normally when the CRT has to start. As a conclusion less patients need feeding tubes during radiottherapy. Last but not least, most patients die of systemic disease and we hope to better target microscopic disease with induction chemotherapy, however, this has not yet be proven.

Oncol Res Treat 2015;38(suppl 5):1–270

115


CONTENTS AUTHOR INDEX

For this reason we are eagerly awaiting new trials with specific focus on induction chemotherapy. Disclosure: No conflict of interest disclosed.

3 Blom RL et al.: Dis Esophagus 2014;27:380–387. 4 van Hagen P et al.: N Engl J Med 2012;366:2074–2084. 5 Honing J et al.: Ann Oncol 2014;25:638–643. Disclosure: No conflict of interest disclosed.

V405

Inductionchemotherapy prior to radiochemotherapy and operation in esophageal cancer – the CONTRA position

Fortbildung

Patientensicherheit

Thuss-Patience P.1 Charite Universitätsmedizin Berlin, Med. Klinik m S Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany 1

Due to the dismal prognosis of esophageal cancer we feel the need to maximize therapy as long as possible with the hope to also maximize efficacy. The rationale for induction-chemotherapy prior to radiochemotherapy and operation is based on the hope to maximize the effect on micrometastases which are not covered in the radiation field. In my talk I will critically check whether there are any reliable data which support this feeling and whether induction chemotherapy really translates into any clinical benefit for the patient. Disclosure: Peter Thuss-Patience: Advisory Role: Roche, Lilly, Nordic; Expert Testimony: GSK. V406

What is the optimal chemotherapy for neoadjuvant and definitive chemoradiotherapy? Eisterer W.1 Medizinische Universität Innsbruck, Univ.-Klinik für Innere Medizin I, Internistische Onkologie, Innsbruck, Austria 1

With an increasing incidence and overall 5-year survival of about 15%, the prognosis of esophageal cancer (EC) patients remains poor. Neoadjuvant chemoradiation (nCRT) has shown to be superior compared with surgery alone leading to the current standard procedure in medically fit esophageal carcinoma with curative resectable EC. In patients who are not eligible for curative intended surgery due to technical reasons or medically unfit for resection definitive chemoradiotherapy (dCRT) respresents an alternative option. The most commonly used regimens are those consisting of cisplatinum in combination with 5-fluorouracil (5FU) or paclitaxel combined with carboplatin. Current guidelines in the United States and Europe recommend the combination of cisplatinum with 5-FU as standard combined with 50.4 Gy radiation therapy [1] while the carboplatin/paclitaxel regimen is frequently used in patients with extensive comorbidity [2]. A comparison of two nCRT regimens in patients with potentially curable EC proved that the carboplatin/paclitaxel/41.4 Gy regimen caused less toxicity compared with the cisplatin/5-FU/50.4 Gy regimen, with an insignificant difference in response rates and long-term survival [3]. In the Dutch CROSS Trial [4] paclitaxel and carboplatin weekly for 5 weeks with 41.4 Gy radiotherapy led to a significant survival benefit compared to surgery alone; nCRT resulted in 23% pathologic complete Response, less locoregional relapse and no compromise of the surgical resection. A retrospective analysis of dCRT with carboplatin/Paclitaxel versus cisplatin/5FU in 102 patients [5] showed similar survival between the two regimes. Carboplatin/paclitaxel was completed more frequently (82% vs 57%) and showed less hematologic and non-hematologic toxicity (grade 3 or higher 4% vs 18%). Conclusion: Carboplatin/paclitaxel represents a valid alternative to cisplatin/5FU. Further development of taxane-based CRT schedulesare warranted. References: 1 Stahl M et al.: Ann Oncol 2010; 21(Suppl 5):v462. 2 National Comprehensive Cancer Network. Clinical Practical Guidelines in Oncology. Esophageal and Esophagogastric Junction Cancers Version 2.2013. 2013:ESOPH-E 6.-v49.

116

Oncol Res Treat 2015;38(suppl 5):1–270

V408

Medication safety from a pharmaceutical perspective Jaehde U.1 Institute of Pharmacy, University of Bonn, Clinical Pharmacy, Bonn, Germany

1

Cancer patients are exposed to a variety of therapy-associated risks when treated with anticancer drugs. Due to the high toxicity and complexity of most therapeutic regimens there is a high incidence of drug-related problems such as adverse effects, drug-drug interactions, and non-adherence. Therefore, there is a strong need for specific interventions assuring medication safety. The provision of a structured medication management is a novel approach to reduce the incidence of adverse events and enhance medication adherence. In our working group, we have developed several interventions to increase the medication safety of cancer patients. For example, we have shown that a structured medication management can reduce the incidence of nausea and emesis in patients with breast and ovarian cancer [1]. Moreover, a prospective two-arm cohort study revealed the potential of an adherence management for patients receiving the orally administered anticancer drug capecitabine [2]. Recently, we have developed a novel modular medication management for outpatient cancer care in a multiprofessional quality circle consisting of oncologists, pharmacists and nurses defining the tasks and responsibilities of each profession. All care modules include evidence-based recommendations for supportive care, written patient information, and an algorithm illustrating the care process. Care modules were developed for medication review and interaction check, malnutrition, and for the management of four common adverse events: nausea/emesis, mucositis, fatigue, and pain. They can be applied individually for each patient according to medication and anticipated toxicity. The feasibility of the model has recently been evaluated in a randomized two-arm interventional trial [3]. In conclusion, a structured medication management can reduce the incidence of drug-related problems and hence improve medication safety. Multiprofessional approaches, including at least oncologists, pharmacists and nurses, are most promising with regard to implementation in daily routine. References: 1 Liekweg A, Westfeld M, Braun M, Zivanovic O, Schink T, Kuhn W, Jaehde U. Support Care Cancer 2012;20:2669–2677. 2 Krolop L, Ko YD, Schwindt PF, Schumacher C, Fimmers R, Jaehde U. BMJ Open 2013;3:e003139. 3 Wilmer A, Jansen C, Ko YD, Schmidt-Wolf I, Jaehde U. FORUM 2014;29:324–330. Disclosure: No conflict of interest disclosed. V410

Learning from errors: Clinical risk management in the oncology Paula H.1 Inselspital, Universitätsspital Bern, Ärztliche Direktion, Qualitätsmanagement, Bern, Switzerland 1

Introduction: Learning from errors is one of the most common principles in the clinical risk management. Indeed it is undisputable important to detect and understand the reasons why things are going wrong. Especially in high-risk disciplines the knowledge about these facts is the key to avoid

Abstracts


CONTENTS AUTHOR INDEX

adverse events. Usually in hospitals the emergency medicine, operating theatres, anaesthesia and intensive care are reckoned to this category. The hazardous nature of oncology is frequently underestimated in this context. Highly potential therapies and vulnerable patients are a very dangerous mixture. Therefore it is no exaggeration to declare oncology also as a high-risk discipline. Methods: In health care the most activities are based on human decisions and performances. In the case of an adverse event it is thus on the first sight mostly no problem to identify one or more persons which have caused an error. This leads into ignoring other important facts. At a closer look it is feasible to detect error promoting elements in the environment or in the organisational structure. There are a few established and approved methods to achieve this goal. More than mortality and morbidity conferences or root-cause-analyses incident reporting is widely-used. CIRS (Critical Incident Reporting System) means to collect incidents, which have caused no harms (“near misses”) with the purpose of the identification of error promoting factors. Results: Despite of the worldwide distribution of incident reporting systems a lot of the referred problems are still unresolved. Theoretically enough information are available to manage the reported risks. In reality there are still a lot of unanswered questions. One factor might be the conventional way of interpreting critical incidents. Perhaps it is more promising not to ask what was going wrong, but to ask which factors are usually responsible for an error-free treatment. Conclusions: In the foreseeable future incident reporting remains as an important tool. However, the system should be critically scrutinized on existing opportunities for improvements. One possible approach is to focus on factors that are normally ensuring the error free treatment. This so called “Safety II” (according to Hollnagel et al) might be a useful strategy to enhance patient safety. It is quite possible to use CIRS as an information source in this context. But it is necessary to direct the focus on other facts and to ask questions that never have been asked before. Disclosure: No conflict of interest disclosed.

Fortbildung

Hämostaseologie II V412

Screening for occult cancer after idiopathic VTE – not too much and not too little Matzdorff A.1 Asklepios Klinikum Uckermark, Mediz. Klinik II, Hämatologie, Onkologie, Schwedt, Germany 1

Cancer can trigger thromboembolism. There is a 4–10% chance of finding an asymptomatic occult cancer in patients with idiopathic venous thromboembolism. So far guidelines recommend limited cancer screening with history taking, physical examination, and screening examinations as recommended by age. Studies show that a more extensive screening program, including endoscopy and computed tomography finds more cancers at an early stage with a better chance of cure. While extensive cancer screening detects more cancers it has, as of yet, not been shown to reduce overal mortality. More arguments against extensive cancer screening are the increased risk of false-positive results, which lead to additional diagnostic interventions with more side effects and complications and higher costs. It is highly unlikely that there will be another randomized study on limited vs. extensive cancer screening large enough to show an effect on mortality or cost-effectiveness. Considering the clinical need for advice and guidance on this topic and the consequences for the individual patient when missing a cancer diagnosis the following screening program for patients with idiopathic VTE is suggested: 1. Medical history including risk factors for cancer, 2. Physical examination including rectal exam,

Abstracts

3. Complete blood count, electrolytes, liver and renal function tests, urinalysis, and with atypically localized thromboses (e.g. abdominal vein thromboses) also JAK2 and CALR mutation screening, 4. Women: breast exam*, mammography*, pelvic exam*, PAP-smear*, 5. Men: prostate exam*, transrectal ultrasound*, PSA* (PSA never alone, always combined with the aforementioned exams), 6. Tumormarker-Screening (CEA, CA 12–5, αFP, never alone, only following positive findings from other studies to narrow the differential diagnosis), 7. New recommendation: gastroscopy and colonoscopy*, 8. New recommendation: CT or – if available – MRI of abdomen and pelvis, 9. New recommendation: low-dose chest CT, (* if not already received as part of prior cancer screening.) The pros and contras of this approach will be discussed in the presentation. Disclosure: No conflict of interest disclosed. V413

Anticoagulation in liver cirrhosis Langer F.1 Universitätsklinikum Eppendorf, II. Medizinische Klinik und Poliklinik, Hamburg, Germany 1

The coagulopathy of liver cirrhosis is characterized by downregulation of both procoagulant (e.g. fibrinogen and clotting factors II, V, VII and X) and anticoagulant factors such as antithrombin (AT), protein C and protein S. Consequently, even under conditions of severe hepatic dysfunction, the hemostatic system in patients with chronic liver disease is typically balanced with regard to pro- and anticoagulant pathways. Commonly used coagulation tests such as the prothrombin time (PT) and the activated partial thromboplastin time (APTT), however, do not assess the function of coagulation inhibitors. In particular, the PT is not influenced by a defect in the activated protein C pathway, and the spontaneously prolonged PT in patients with liver cirrhosis has therefore long been interpreted as a laboratory sign of systemic hypocoagulability. More global coagulation tests such as calibrated automated thrombography (CAT) in the presence of soluble thrombomodulin, however, have clearly shown that thrombin generation in plasmas from cirrhosis patients is not significantly decreased when activation of the protein C system is enabled. In fact, plasma thrombin generation capacity may even be increased in patients with liver cirrhosis, and vascular thrombotic events are increasingly recognized in this patient population. Although cirrhosis patients may experience severe bleeding symptoms, mainly as a result from portal hypertension, endothelial dysfunction, infection and/or uremia, the coagulopathy of liver cirrhosis in itself does not preclude systemic anticoagulation, especially, since accompanying thrombocytopenia is typically counterbalanced by massively increased plasma levels of von Willebrand factor. The use of vitamin K antagonists (VKAs) in cirrhosis patients is hampered by the spontaneously prolonged PT, which is partially due to clotting factor V deficiency. The INR may therefore not adequately reflect the antithrombotic effects of VKAs. In this regard, the direct oral anticoagulants rivaroxaban, apixaban and dabigatran may have several pharmacological advantages, but their use in patients with severe hepatic and/ or renal dysfunction is limited. For this reason, low-molecular heparins still play a major role in the prevention and treatment of thromboembolic events in cirrhosis patients, but sufficiently high AT plasma levels are required for these indirect agents to exert there anticoagulant effects. Disclosure: Florian Langer: Advisory Role: Baxter, Bayer, Bristol-Myers Squibb, CSL Behring, Daiichi-Sankyo, LEO, Novartis, Novo Nordisk, Pfizer; Financing of Scientific Research: Baxter, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, CSL Behring, Daiichi-Sankyo, LEO, Novartis, Novo Nordisk, Pfizer, Sanofi; Expert Testimony: CSL Behring, Pfizer.

Oncol Res Treat 2015;38(suppl 5):1–270

117


CONTENTS AUTHOR INDEX

Freier Vortrag

Conclusions: In pts with previously untreated indolent lymphomas, and in elderly pts with MCL, B-R demonstrates a PFS and TTNT benefit over CHOP-R. Additionally, treatment with B-R resulted in a trend toward a survival benefit in the group of pts with indolent lymphomas.

Lymphome indolent V418

Bendamustine plus Rituximab (B-R) versus CHOP plus Rituximab (CHOP-R) as first-line treatment in patients with indolent lymphomas or mantle cell lymphomas (MCL) – 7-year updated results from the StiL NHL1 study Rummel M.1, Maschmeyer G.2, Ganser A.3, Heider A.4, von Grünhagen U.5, Losem C.6, Heil G.7, Welslau M.8, Balser C.9, Kaiser U.10, Weidmann E.11, Dürk H.12, Balló H.13, Stauch M.14, Barth J.1, Blau W.1, Burchardt A.1, Kauff F.1, Brugger W.15, StiL (Studiengruppe indolente Lymphome) Universitätsklinik, Med Kl IV, Hämatologie, Gießen, Germany, 2Ernst von Bergmann Klinikum, Potsdam, Germany, 3Medizinische Hochschule Hannover, Hannover, Germany, 4Klinikum Leverkusen gGmbH, Leverkusen, Germany, 5 Schwerpunktpraxis, Cottbus, Germany, 6Facharztzentrum, Neuss, Germany, 7 Klinikum Lüdenscheid, Lüdenscheid, Germany, 8Phase drei, HämatoOnkologischer Studienkreis am Klinikum Aschaffenburg, Aschaffenburg, Germany, 9Gemeinschaftspraxis, Marburg, Germany, 10St. Bernward Krankenhaus, Hildesheim, Germany, 11Krankenhaus Nordwest, Frankfurt/ Main, Germany, 12St. Marien-Hospital Hamm gGmbH, Hamm, Germany, 13 Gemeinschaftspraxis, Offenbach, Germany, 14Hämatologisch-Onkologische Schwerpunktpraxis, Kronach, Germany, 15Schwarzwald-Baar Klinikum, VillingenSchwenningen, Germany 1

Introduction: This multicenter, randomized, phase III study compared B-R and CHOP-R as first-line treatment in patients (pts) with indolent lymphomas or MCL and was published in The Lancet in 2013. The final published analysis at a median follow-up of 45 months demonstrated a significantly prolonged progression-free survival (PFS) in the B-R group, compared to the CHOP-R group (p < 0.001). Median PFS was 69.5 vs. 31.2 months, respectively. In the current analysis, we present updated results for overall survival (OS), time-to-next-treatment (TTNT), and secondary malignancies (sNPL) with a median follow-up of 87 months. Methods: 549 pts with indolent lymphomas or MCL were randomized to receive B-R or CHOP-R for a maximum of 6 cycles. The primary endpoint was PFS; secondary endpoints included OS, TTNT, and sNPL. Results: 514 randomized pts were evaluable (261 B-R; 253 CHOP-R). Patient characteristics were well balanced between arms; median age was 64 years. Fewer pts treated initially with B-R needed second-line treatments due to disease progression compared to CHOP-R treated pts: 93 pts (36%) in the B-R group received salvage treatment compared with 140 pts (55%) in the CHOP-R group. Of those in the CHOP-R group, 69 pts (49%) received B-R as salvage. TTNT was significantly prolonged with B-R compared with CHOP-R (p < 0.001). Median TTNT was not yet reached in the B-R group vs. 42.3 months in the CHOP-R group. The difference in complete response (CR) rates (independent of treatment arms) between male (n = 272, median age 63 years) and female (n = 242, median age 64 years) pts was statistically significant: 28.6% for male pts versus 42.1% for female pts (p = 0.0016). Female pts had a longer median TTNT compared to male pts (not yet reached vs. 52.2 months, respectively; p = 0.006). The achievement of a CR was associated with significantly prolonged OS, with an estimated 10-year survival rate of 72.6% for pts with a CR and 63.6% for pts with a partial response (p = 0.006). The difference in OS between the treatment arms was not statistically significant. The estimated 10-year survival rates were 67.4% for B-R and 60.1% for CHOP-R (p = 0.262). In pts with indolent lymphomas (without MCL), there was a trend toward a longer survival for the B-R group compared with the CHOP-R group, with 43/215 pts (20.0%) in B-R and 58/205 pts (28.3%) in CHOP-R. The estimated 10-year survival rates for pts with indolent lymphomas were 71.9% for B-R and 61.5% for CHOP-R (p = 0.076). No difference in OS was found in the subgroup of pts with MCL (n = 95; p = 0.429). Twenty sNPL were observed in the B-R group compared with 23 in the CHOP-R group, with 1 hematological malignancy in each group to date.

118

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: Mathias Rummel: Financing of Scientific Research: Amgen, Celgene, Gilead, Janssen, Mundipharma, Roche; Expert Testimony: Grifols, Mundipharma, Roche. Wolfram Brugger: Advisory Role: Ja; Financing of Scientific Research: ­Mundipharma, Roche Pharma; Other Financial Relationships: Teilnahme an wissenschaftlichen Kongressen. V419

Bendamustine plus Rituximab versus Fludarabine plus Rituximab in patients with relapsed follicular, indolent, or mantle cell lymphomas – 8-year follow-up results of the randomized phase III study NHL 2–2003 on behalf of the StiL (Study Group Indolent Lymphomas, Germany) Rummel M.1, Balser C.2, Kaiser U.3, Balló H.4, Stauch M.5, Heider A.6, Welslau M.7, Losem C.8, Weidmann E.9, Blau W.1, Burchardt A.1, Barth J.1, Kauff F.1, Brugger W.10, StiL (Studiengruppe indolente Lymphome) Universitätsklinik, Med Kl IV, Hämatologie, Gießen, Germany, Gemeinschaftspraxis, Marburg, Germany, 3St. Bernward Krankenhaus, Hildesheim, Germany, 4Gemeinschaftspraxis, Offenbach, Germany, 5 Hämatologisch-Onkologische Schwerpunktpraxis, Kronach, Germany, 6 Klinikum Leverkusen gGmbH, Leverkusen, Germany, 7Phase drei, HämatoOnkologischer Studienkreis am Klinikum Aschaffenburg, Aschaffenburg, Germany, 8Facharztzentrum, Neuss, Germany, 9Krankenhaus Nordwest, Frankfurt/Main, Germany, 10Schwarzwald-Baar Klinikum, VillingenSchwenningen, Germany 1 2

Introduction: No standard exists for patients (pts) with relapsed/refractory follicular lymphoma (FL), other indolent lymphoma (oiNHL), or mantle cell lymphoma (MCL). Treatment with fludarabine containing regiments were common. In 2003 we initiated a multicenter, randomized phase III study to compare the efficacy and safety of bendamustine plus rituximab (B-R) versus F-R for pts with relapsed FL, oiNHL or MCL to further improve the treatment. Methods: 230 pts with relapsed FL, oiNHL, or MCL in need of treatment were randomized to rituximab 375 mg/m² (day 1) plus either bendamustine 90 mg/m² (days 1+2) or fludarabine 25 mg/m² (days 1–3) q 28 days for a maximum of 6 cycles. Prophylactic use of antibiotics or G-CSF was not generally recommended; however, in cases of severe granulocytopenia, G-CSF use was permitted. The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), overall response rate (ORR), and complete response rate (CR). Results: A total of 219 pts were evaluable for the analysis (114 B-R; 105 F-R). There were no significant differences between arms for patient characteristics. Most pts had stage IV (71.6% B-R; 60.6% F-R) or stage III disease (21.1% B-R; 25.3% F-R). Median pts age was 68 yrs (range 38–87). Pts had received a median of 1 prior therapy (range 1–7). Histological subtypes were distributed equally between the B-R and F-R arms. A median of 6 cycles were given in both treatment arms, with 75.2% and 53.4% of B-R and F-R pts receiving 6 cycles, respectively. At the time of this analysis, the median observation time was 96 months. The ORR was significantly higher with B-R than with F-R (83.5% vs. 52.5%, respectively; p < 0.0001). The CR rate with B-R was also significantly higher than that with F-R (38.5% vs. 16.2%; p = 0.0004). Median PFS was significantly prolonged with B-R compared with F-R (34 vs. 12 months p < 0.0001). The longer PFS translated into a survival benefit with a significantly longer median overall survival in the B-R group than in the F-R group (110 vs. 49 months; p = 0.0125) comprising 55 and 71 deaths in the B-R and F-R groups, respectively. There were no significant differences in the rates of alopecia, stomatitis, erythema, allergic reactions, peripheral neuropathy, or infectious episodes between groups. Hematologic toxicities were also similar between arms. The overall incidence of serious adverse events was similar for the B-R and F-R groups (17.4% and 22.2%, respectively). 17 pts (14.9%) developed a

Abstracts


CONTENTS AUTHOR INDEX

secondary neoplasia after B-R compared with 16 pts (15.2%) after F-R. Of these, 5 pts in the B-R group, and 3 pts in the F-R group developed a secondary hematological neoplasia. Conclusions: B-R was more effective than F-R in this setting of relapsed FL, other indolent lymphomas and MCL due to higher overall and complete response rates, a longer PFS, and an improved OS. These data confirm the high anti-lymphoma activity of B-R. Disclosure: Mathias Rummel: Financing of Scientific Research: Amgen, Celgene, Gilead, Janssen, Mundipharma, Roche; Expert Testimony: Grifols, Mundipharma, Roche. Wolfram Brugger,: Advisory Role: Ja; Financing of Scientific Research: ­Mundipharma, Roche Pharma; Other Financial Relationships: Teilnahme an wissenschaftlichen Kongressen. V420

Treatment of refractory hairy cell leukemia by BRAF-inhibition Dietrich S. , Pircher A. , Andrulis M.A. , Peyrade F. , Wendtner C.-M. , Gaehler A.6, Follows G.7, Dyer M.8, Elter T.9, Zeiser R.10, Herrmann M.11, Herold M.11, Dearden C.12, Haferlach T.13, Hallek M.14, Hüllein J.15, Matutes E.12, Dürig J.16, von Kalle C.15, Glimm H.15, Fröhling S.15, AbdelWahab O.17, Hutter B.18, Steurer M.2, Ho A.3, Zenz T.15 1

2

3

4

5

Universitätsklinik Heidelberg, Medizinsiche Klinik V, Heidelberg, Germany, Universität, Innsbruck, Austria, 3Universitätsklinik, Heidelberg, Germany, 4 University, Nice, France, 5Krankenhaus Schwabing, München, Germany, 6 Kantonspital, Luzern, Switzerland, 7University of Cambridge, Cambridge, United Kingdom, 8University of Leicester, Leicester, United Kingdom, 9 Universitätsklinikum, Köln, Germany, 10Universitätsklinikum, Freiburg, Germany, 11 Helios Klinikum, Erfurt, Germany, 12Royal Marsden Hospital, London, United Kingdom, 13MLL Münchner Leukämie Labor, München, Germany, 14Universität, Köln, Germany, 15NCT, Heidelberg, Germany, 16Universitätsklinikum, Essen, Germany, 17MSKCC, New York, United States, 18DKFZ, Heidelberg, Germany 1 2

Background: An activating mutation of the BRAF serine/threonine protein kinase (BRAF V600E) is the key genetic lesion in hairy cell leukemia patients (HCL), suggesting oncogene dependence and opportunities for therapeutic targeting. Methods: We have collected and analyzed clinical course, molecular studies and follow up of 21 HCL patients treated with the BRAF inhibitor vemurafenib. In three of these patients we performed whole exome sequencing to better understand treatment refractory disease course. Results: BRAFi treatment led to improved blood counts and clinical response in all patients. The overall response rate (ORR) was 95% with 8 CRs (38%) and 12 PRs (57%). A forth of the standard dose of vemurafenib was sufficient (240 mg bid, n = 12/21) to completely abrogate p-ERK signaling of hairy cells in the bone marrow, including patients with PR. Time to blood count improvement (e.g. median time to platelet > 100/ nl 28 days) and decrease of sCD25 serum levels (80% within the first two weeks) was very fast and no difference between patients with low (≤240 mg bid) and higher vemurafenib doses was observed. Off drug relapses occurred in 11/21 patients including patients with CR or treated upfront with vemurafenib after a median of 17 months since treatment cessation. Nine patients were retreated and responded to a second course of BRAFi (n = 6) and monoclonal antibodies with and without purine analogues (n = 3). To better understand disease biology and course of HCL treated with BRAFi, we performed whole exome sequencing in 3 patients. In addition to the disease defining BRAFV600E mutations, we identify EZH2, ARID1A mutations and recurrent inactivating mutations of the cell cycle inhibitor CDKN1B (p27). Targeted deep sequencing of CDKN1B in a larger cohort of HCL patients identify deleterious CDKN1B mutations in 16% of patients with HCL (n = 13/81). In 11 of 13 patients the CDKN1B mutation was clonal, implying an early role of CDKN1B mutations in the pathogenesis of HCL. Conclusion: Our data provide clinical proof of the critical dependence on active BRAF signaling in HCL. We provide conclusive evidence that anti-tumor effect and side effects are observed at fractions of the standard vemurafenib dosing, but off drug relapses occur inevitably. Exome

Abstracts

sequencing reveals that additional cooperating recurrent genetic events occur and may contribute to the disease biology of HCL. Disclosure: No conflict of interest disclosed. V421

Advanced stage follicular lymphomas with and without breaks in the BCL2 locus do not differ with respect to clinical features and outcome Leich E.1, Unterhalt M.2, Wartenberg M.3, Siebert R.4, Klapper W.5, Engelhard M.6, Stuhlmann-Laeisz C.5, Koch K.5, Puppe B.7, Horn H.8,9,10, Bernd H.-W.11, Feller A.C.11, Hummel M.12, Lenze D.12, Stein H.13, Hartmann S.14, Hansmann M.L.14, Möller P.15, Hiddemann W.2, Dreyling M.2, Hoster E.16, Ott G.17, Rosenwald A.3, German Low Grade Lymphoma Study Group Institute of Pathology,University of Würzburg, Würzburg, Germany, Department of Internal Medicine III, University Hospital, Munich, Germany, 3 Institute of Pathology, University of Würzburg, Würzburg, Germany, 4Institute of Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Kiel, Germany, 5Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Kiel, Germany, 6Department for Radiotherapy, University Hospital, Essen, Germany, 7Pathologisches Institut/Universität, Würzburg, Germany, 8Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany, 9 Robert Bosch Krankenhaus, Stuttgart, Germany, 10University, Tuebingen, Germany, 11Institute of Pathology, University Hospital Schleswig-Holstein, Lübeck, Germany, 12Institute of Pathology, Campus Benjamin Franklin, Charité Universitätsmedizin, Berlin, Germany, 13Pathodiagnostik, Berlin, Germany, 14 Institute of Pathology, University Hospital, Frankfurt a.M., Germany, 15Institute of Pathology, University Hospital, Ulm, Germany, 16Institute of Medical Informatics, Biometry, and Epidemiology, University of Munich, Munich, Germany, 17Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany 1 2

Introduction: Approximately 15% of follicular lymphomas (FL) lack chromosomal breaks in BCL2. Clinical features of this subgroup remain largely undefined. The aim of this study, therefore, was to improve knowledge on molecular and clinical features of breakpoint negative FL. Participants and Methods: We studied the presence or absence of chromosomal breaks in BCL2, BCL6 and MYC genes by fluorescence in situ hybridization in large clinical trial cohorts of FL stages III/IV (n = 540) and I/II (n = 117) patients enrolled in the German Low Grade Lymphoma Study Group trials, treated with chemotherapy regimens with or without rituximab and radiation, respectively. The protein expression status of BCL2, BCL6, MUM1, CD10 and P53 and the proliferation status of Ki67 was determined by immunohistochemistry. Finally, we compared the biological and clinical features of FL grades 1–3A with and without chromosomal breaks in BCL2. Results: Chromosomal breaks in BCL2 were detected in 86% of FL stages III/IV and in 53% of FL stages I/II. BCL2 was expressed in almost all t(14;18)-positive FL and surprisingly also in 86% and 69% of t(14;18)-negative FL stages III/IV and I/II, respectively. CD10 expression was significantly reduced in BCL2-break negative FL of all stages. However, there was high similarity between FL with and without BCL2-break with respect to the other molecular features and the clinical parameters investigated. Most importantly, median overall survival and time to treatment failure did not differ significantly between stage III/IV FL with and without breaks in BCL2. Likewise, no significant difference for FLIPI score and performance-status was observed. Conclusion: The results of this large retrospective study suggest that FL with and without breaks in BCL2 show a high degree of similarity with regard to most molecular markers investigated and in particular with regard to their clinical behaviour. This supports the notion that FL with and without breaks in BCL2 represent two subgroups within the same entity. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

119


CONTENTS AUTHOR INDEX

V422

V423

Gastric marginal zone B-cell lymphoma of the mucosaassociated lymphoid tissue (MALT lymphoma) in the course of time: The Vienna University experience

Treatment of low grade B-cell-lymphoma cell lines with arylindolylmaleimide derivates PDA-66 and PDA-377 induces antiproliferative effects

Kiesewetter B.1, Dolak W.2, Wöhrer S.3, Mayerhoefer M.E.4, Simonitsch-Klupp I.5, Chott A.6, Jäger U.7, Raderer M.1

Eschenburg A.1, Roolf C.1, van der Wall K.1, Sklarz L.-M.1, Jaenecke C.1, Sekora A.1, Murua Escobar H.1, Rolfs A.2,3, Pews-Davtyan A.4, Beller M.4, Junghanss C.1

Medizinische Universität Wien / Universitätsklinik für Innere Medizin I, Onkologie, Wien, Austria, 2Medizinische Universität Wien / Universitätsklinik für Innere Medizin III, Gastroenterologie und Hepatologie, Wien, Austria, 3 Medizinische Universität Wien / Universitätsklinik für Innere Medizin I, Knochenmarktransplantation, Wien, Austria, 4Medizinische Universität Wien, Radiologie, Wien, Austria, 5Medizinische Universität Wien, Pathologie, Wien, Austria, 6Wilhelminenspital, Pathologie und Mikrobiologie, Wien, Austria, 7 Medizinische Universität Wien / Universitätsklinik für Innnere Medizin I, Hämatologie und Hämostaseologie, Wien, Austria 1

Introduction: Gastric MALT lymphoma, most prominent for its association with Helicobacter pylori (HP), has been well characterized in the last three decades. However, changes in clinicopathological features and incidence have recently been suggested by scattered reports. Methods: We report our experience of 320 consecutive MALT lymphoma patients diagnosed and followed at our institution 1999–2015. Patients with gastric lymphoma were reassessed for clinicopathological characteristics including treatment and outcome. Results: 116/320 patients (36%) were of gastric origin, while the majority of patients had extragastric MALT lymphoma. Only 4/116 patients (3%) showed an additional diffuse-large B-cell lymphoma component. Using a standardized staging protocol we found 25 patients (22%) to have disseminated disease, with only 3 (3%) having bone marrow involvement, while 78% had localized disease. Out of 99 patients with a fully tested HP-status, 34 (34%) were negative. Interestingly, 9 of them had been diagnosed before 2004 (9/34, 26%) and 25 between 2004–2015 (74%). 40% of patients had monoclonal gammopathy and plasmacytic differentiation was present in 31%. In addition, 29% were found to have an underlying autoimmune disease, with chronic thyroiditis being most common. In terms of treatment, 76 patients (66%) had antibiotics as first line therapy, 31 systemic therapy (27%), while the remaining had surgery, radiation or wait and see. After an impressive median FUP of 63m, 85% (99/116) are alive, while 17 (15%) have died. HP-status was not associated with relapse after first line therapy (p = 0.07), and there was no difference in time to progression between the cohorts. 14 HP-negative patients were treated with antibiotics alone, resulting in 5 CR and one PR (46%), 5 SD and 3 progressions. Results of antibiotic treatment in HP negative patients were not significantly different for time to progression (p = 0.1) and progression following first line (p = 0.38). Transformation to DLBCL was only seen in one patient while two developed clonally unrelated DLBCL. Conclusion: Our findings suggest, that the rate of extragastric MALT lymphoma appears to be higher than stated in the literature and might have been underestimated. In addition, cautious interpretation of our data proposes an increase of HP-negative gastric lymphoma over the years and that also this collective can be managed safely with antibiotics. Our data also show that the risk of DLBCL transformation is minimal.

Universitätsmedizin Rostock, Klinik III, Hämatologie, Onkologie, Palliativmedizin, Rostock, Germany, 2Universitätsmedizin Rostock, AlbrechtKossel-Institute for Neuroregeneration, Rostock, Germany, 3Centogene AG, Rostock, Germany, 4University of Rostock, Leibniz-Institute for Catalysis, Rostock, Germany 1

Introduction: B-cell lymphomas are the most common type of non-Hodgkin lymphomas (NHL). Curative options for indolent lymphoma patients diagnosed in advanced stages are currently limited. The novel arylindolylmaleimide PDA-66 has been shown to inhibit the polymerisation of microtubules inducing antiproliferative effects in acute lymphoblastic leukemia cells. Here, we evaluated the effects and potency of PDA-66 and its derivate PDA-377 on B-cell NHL cell lines. Methods: NHL cell lines (SC-1, MINO and U-266) were treated with increasing concentrations of PDA-66 and PDA-377 (0.5 µM-5 µM) and incubated for 72 h. Metabolic activity was determined by WST-1 assay. Furthermore, gene expression changes were investigated after 2 and 24 h treatment with PDA-66 using human signal transduction pathway finder qPCR assay. Further, 1 µM PDA-66 was tested in combination with 25 nM cytarabine (AraC), 50 nM doxorubicin (Doxo) or 10 nM dexamethasone (Dexa) on SC-1 cells, all in IC-80 concentrations. The applied cytostatics were added either at the time of cell seeding simultaneously with PDA-66, or 24 h before, or after PDA-66 treatment, respectively. Results: Metabolic activity was significantly reduced in a dose dependent manner in all cell lines starting at a concentration of 1 µM PDA-66 and 2.5 µM PDA-377. Treatment with 5 µM PDA-66 decreased metabolic activity to 34.4% (SC-1), 7.9% (MINO) and 29.7% (U-266) compared to control cells treated with DMSO (100%). Incubation with 5 µM PDA-377 reduced metabolic activity to 39.4% (SC-1), 51.1% (MINO) and 40.5% (U-266) respectively. Gene expression analysis showed that PDA-66 influenced NFkB pathway by down regulation of TNF gene. Key genes of other investigated pathways like Wnt, JAK/STAT, p53 and NOTCH were not affected. Treatment with PDA-66 demonstrates synergistic effects on proliferation inhibition when combined with AraC, Doxo or Dexa using Bliss statistics. The observed combined effect was independent from PDA-66/cytostatic application showing no enhanced impact on the metabolic activity. Conclusions: Our data indicates that application of PDA-66 alone or in combination with cytostatic drugs shows distinct effects on B-NHL cells impacting the NFkB signaling pathway. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

120

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag AML klinisch V424

Prolonged survival in patients ≥60 years with first relapsed/ refractory acute myeloid leukemia treated with vosaroxin plus cytarabine vs placebo plus cytarabine: Results from the phase 3 VALOR study Krug U.1, Ravandi F.2, Ritchie E.K.3, Sayar H.4, Lancet J.E.5, Craig M.D.6, Vey N.7, Strickland S.A.8, Schiller G.J.9, Jabbour E.2, Erba H.P.10, Pigneux A.11, Horst H.-A.12, Recher C.13, Klimek V.M.14, Cortes J.2, Roboz G.J.3, Craig A.R.15, Fox J.A.15, Ward R.15, Smith J.A.15, Acton G.15, Mehta C.16, Kantarjian H.M.2, Stuart R.K.17 Universitätsklinikum Münster, Leverkusen, Germany, 2University of Texas MD Anderson Cancer Center, Houston, United States, 3Weill Cornell Medical Center, New York, United States, 4Indiana University Cancer Center, Indianapolis, United States, 5Moffitt Cancer Center, Tampa, United States, 6West Virginia University, Morgantown, United States, 7Institut Paoli-Calmettes and AixMarseille University, Marseille, France, 8Vanderbilt-Ingram Cancer Center, Nashville, United States, 9University of California, Los Angeles, United States, 10 University of Alabama, Birmingham, United States, 11Université de Bordeaux, CHU de Bordeaux, Bordeaux, France, 12II. Medizinische Klinik und Poliklinik im Städtischen Krankenhaus, Kiel, Germany, 13Institut Universitaire du Cancer de Toulouse Oncopole, Université de Toulouse III, CHU de Toulouse, Toulouse, France, 14Memorial Sloan-Kettering Cancer Center, New York, United States, 15 Sunesis Pharmaceuticals, Inc., South San Francisco, United States, 16Cytel, Inc. and Harvard School of Public Health, Cambridge, United States, 17Medical University of South Carolina, Charleston, United States 1

Fig. 1. Krug VALOR Bild. Disclosure: Utz Krug: Advisory Role: Sunesis Pharmaceuticals, Roche, Gilead Sciences, Novartis, Catapult Cell Therapeutics, Celgene, Clavis Pharma; Expert Testimony: Boehringer Ingelheim; Other Financial Relationships: Travel support: Mundipharma, Novartis; data monitoring board fees: German-Austrian AML Study Group. Robert K. Stuart: Advisory Role: Sunesis Pharmaceuticals; Expert Testimony: Sunesis Pharmaceuticals; Other Financial Relationships: Travel support: Sunesis Pharmaceuticals. V425

Introduction: Prognosis for older patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) is poor and treatment options are limited. Vosaroxin is a first-in-class anticancer quinolone derivative active in AML. VALOR, a phase 3, randomized, double-blind, placebo-controlled trial, evaluated vosaroxin plus cytarabine (vos/cyt) vs placebo plus cytarabine (pla/cyt) in patients with R/R AML (NCT01191801). Methods: Patients with refractory disease after primary induction treatment or in first relapse were randomized 1:1 to receive cytarabine (1 g/ m2 IV over 2 h, d 1–5) plus either vosaroxin (90 mg/m2 IV over 10 min d 1 and 4; 70 mg/m2 in subsequent cycles) or placebo. Primary endpoints: overall survival (OS) and 30- and 60-day mortality. Here, we report results of a predefined subgroup analysis in patients ≥60 y of age. Results: Of 711 patients randomized, 63% (n = 451) were ≥60 y (n = 226 randomized to vos/cyt and n = 225 to pla/cyt). At final analysis, OS was improved with vos/cyt (7.1 mo vs 5.0 mo with pla/cyt; HR = 0.75; P = 0.003) (Figure). Complete remission (CR) was achieved in 31.9% of patients treated with vos/cyt vs 13.8% with pla/cyt (P< 0.0001). Responses were durable; median leukemia-free survival in patients who achieved CR was 10.3 mo with vos/cyt vs 6.5 mo with pla/cyt (P = 0.20). Thirty- and 60-day all-cause mortality was similar between treatment arms (30-day: 10.2% vs 9.0%; 60-day: 20.4% vs 22.6% with vos/cyt vs pla/ cyt, respectively). Serious AEs were more common with vos/cyt (57% vs 33% with pla/cyt); most common serious AEs were febrile neutropenia (9.3% with vos/cyt vs 5.9% with pla/cyt), sepsis (9.3% vs 5.0%), pneumonia (7.5% vs 4.5%), bacteremia (7.5% vs 2.3%), and stomatitis (4.4% vs 1.8%). Serious and non-serious cardiac, renal, neurologic, and hepatic AEs were comparable between treatment groups. Conclusions: Addition of vosaroxin to cytarabine improved OS and increased CR rates without increasing early mortality in patients ≥60 y with R/R AML. The toxicity observed in the vosaroxin arm was acceptable in light of the benefit received. VALOR results support the use of vos/cyt as a potential standard of care in patients ≥60 y with R/R AML.

Clinical outcome in older AML patients not fit for intensive chemotherapy: Results of the population-based AMLSG-BiO (NCT01252485) registry study of the German-Austrian AML study group

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Schlenk R.F.1, Paschka P.2, Krauter J.3, Lübbert M.4, Brossart P.5, Salih H.R.6, Theobald M.7, Fiedler W.8, Wulf G.9, Kirchner H.10, Hertenstein B.11, Heidel F.12, Westermann J.13, Grießhammer M.14, Wattad M.15, Götze K.16, Girschikofsky M.17, Kündgen A.18, Koller E.19, Rudolph C.2, Heuser M.20, Thol F.20, Goehring G.20, Teleanu V.2, Weber D.2, Gaidzik V.I.2, Döhner K.2, Ganser A.20, Döhner H.2, GermanAustrian Acute Myeloid Leukemia Study Group (AMLSG) Uniklinik Ulm, Klinik für Innere Medizin III, Ulm, Germany, 2Uniklinik, Ulm, Germany, 3Klinikum, Braunschweig, Germany, 4Uniklinik, Freiburg, Germany, 5 Uniklinik, Bonn, Germany, 6Uniklinik, Tübingen, Germany, 7Uniklinik, Mainz, Germany, 8Uniklinik, Hamburg, Germany, 9Uniklinik, Göttingen, Germany, 10 Klinikum Frankfurt-Höchst, Frankfurt, Germany, 11Klinikum Bremen Mitte, Bremen, Germany, 12Uniklinik, Magdeburg, Germany, 13Charité – Universitätsmedizin, Berlin, Germany, 14Klinikum, Minden, Germany, 15 Klinikum, Essen, Germany, 16Uniklinik München r.d.I, München, Germany, 17 Krankenhaus der Elisabethinen, Linz, Austria, 18Uniklinik, Düsseldorf, Germany, 19 Hanuschkrankenhaus, Wien, Austria, 20Medizinische Hochschule Hannover, Hannover, Germany 1

Background: Comparative population-based outcome data in older AML patients (pts) unfit for intensive chemotherapy (IC) are rare outside interventional treatment trials and treatment options include best supportive care (BSC), decitabine (DAC), azacitidine (AZA), or low-dose cytarabine (LDAC). Aims: To assess outcome in pts >60 years (yrs) unfit for IC within the registry/molecular-screening trial AMLSG BiO (NCT01252485). Methods: A total of 2786 pts (median age 72 yrs, range 60–95) were registered between October 2010 and May 2015. All pts were screened within 48-hours for gene mutations and fusion-genes. Results: Of the 2786 pts, n = 1338 (48%) received IC, n = 107 (4%) AZA, n = 222 (8%) DAC, n = 281 (10%) LDAC, n = 276 (10%) BSC, and n = 562 (20%) unknown treatment. Of 886 non-intensively treated pts, n = 201 (23%) were treated within clinical trials. In the remaining 685 pts treatment was as follows: n = 144 LDAC, n = 162 DAC, n = 106 AZA, and n = 273 BSC. There was no difference between the groups in terms of age

121


CONTENTS AUTHOR INDEX

(p = 0.18, median 76 yrs, range 60–92) and cytogenetic risk groups according to ELN recommendations (p = 0.12; high, 27%; intermediate/low, 73%). Significantly more pts with HCT-CI score >2 were present in the LDAC group (p = 0.03) and with ECOG performance status >1 (p < 0.001) in the BSC group. NPM1 mutations were more frequent (p < 0.001) in BSC and LDAC (22%, each) groups than in AZA (5%) or DAC (7%) group. FLT3-ITD and FLT3-TKD were rarely detected (2.5%, 2.7%, respectively) with no difference between the groups (p = 0.69, p = 0.99). WBC at diagnosis was significantly (p < 0.001) higher in BSC and LDAC (26G/l; each) groups compared to AZA (4.5G/l) or DAC (5.8G/l) groups. According to the 4 groups (BSC, AZA, DAC, LDAC), median survival was 0.9, 10.5, 8.6, and 3.6 months, while one-year and two-year survival rates were 15%, 46%, 39%, 30% and 4%, 15%, 6%, 16%, respectively. In a Cox regression model including the groups AZA, DAC and LDAC, survival was significantly influenced by age [hazard ratio (HR) by 10 yrs difference, 1.46; p = 0.03], high-risk cytogenetics (HR, 1.78; p = 0.003), treatment (reference LDAC) with AZA (HR, 0.39; p < 0.001), DAC (HR, 0.43; p < 0.001), ECOG > 1 (HR, 1.55; p = 0.03) but not by NPM1 (p = 0.70) and FLT3-ITD (p = 0.42) mutational status, and HCT-CI > 2 (p = 0.16). Conclusion: In older AML treatment with AZA or DAC is in the shortterm perspective of one to two years superior to LDAC; results beyond two yrs are still dismal for all treatment options. Disclosure: Richard Schlenk: Advisory Role: IQWIG, Ambit; Financing of Scientific Research: Pfizer, Novartis, Janssen, Celgene, Amgen, Teva; Expert Testimony: Pfizer, Novartis, Janssen, Celgene, Amgen, Teva, Arog. Hartmut Döhner: Advisory Role: Celgene; Financing of Scientific Research: Celgene. V426

DNMT3A mutations in exon 23 are associated with improved survival in AML patients treated with Azacytidine plus standard chemotherapy 2

3

3

1

1

4

Universitätsklinikum, Halle (S.), Germany, 2Klinikum Leverkusen gGmbH, Leverkusen, Germany, 3Uniklinik, Münster, Germany, 4Uniklinik, Dresden, Germany, 5Uniklinik, Aachen, Germany, 6Universität, Bochum, Germany, 7 Uniklinikum, Erlangen, Germany, 8St. Bernward Hospital, Hildesheim, Germany, 9 Uniklinik, Würzburg, Germany, 10Uniklinik, Frankfurt, Germany, 11Uniklinik, Essen, Germany, 12Universitätsklinikum, Mainz, Germany, 13Charité Campus Benjamin Franklin, Berlin, Germany, 14ASKLEPIOS Klinik St. Georg, Hamburg, Germany, 15Klinikum rechts der Isar (Technische Universität München), München, Germany, 16Universitätsklinikum, Regensburg, Germany, 17RobertBosch-Krankenhaus, Stuttgart, Germany, 18Uniklinikum, Marburg, Germany, 19 Klinikum Nuernberg Nord, Nürnberg, Germany, 20Klinikum Chemnitz gGmbH, Chemnitz, Germany, 21Klinikum, Osnabrück, Germany, 22Dr.-HorstSchmidt-Klinik, Wiesbaden, Germany, 23Klinikum, Frankfurt/Oder, Germany, 24 St. Johannes Hospital, Duisburg, Germany, 25Städtische Kliniken, Bielefeld, Germany, 26Klinikum am Bruderwald, Bamberg, Germany, 27Westpfalz-Klinikum, Kaiserslautern, Germany, 28Klinikum, Bayreuth, Germany, 29Stiftungsklinikum Mittelrhein, Koblenz, Germany 1

DNMT3A mutations are among the most common mutations in AML with about half of all mutations found in exon 23 in one hotspot, codon R882. DNMT3A-R882 mutations seem to be associated with specific changes in DNA methylation patterns and worse outcome. A few reports suggest that monotherapy with DNMT inhibitors might be specifically active in AML with DNMT3A mutations. Here, we analyzed the association between DNMT3A mutations and chemotherapy response with or without added azacytidine as DNMT3A inhibitor.

122

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: Petra Tschanter: Financing of Scientific Research: Vortragshonorar von Celgene. Carsten Müller-Tidow: Financing of Scientific Research: Vortragshonorar von Celgene; Expert Testimony: von Celgene und Amgen. V427

Tschanter P. , Krug U. , Füller M. , Klein M. , Göllner S. , Rohde C. , Röllig C. , Thiede C.4, Lilly M.A.4, Haack B.4, Koschmieder A.3, Stelljes M.3, Dugas M.3, Koschmieder S.5, Gerss J.3, Butterfaß-Bahloul T.3, Wagner R.3, Eveslage M.3, Thiem U.6, Krause S.W.7, Kaiser U.8, Kunzmann V.9, Steffen B.10, Noppeney R.11, Herr W.12, Baldus C.D.13, Schmitz N.14, Götze K.15, Reichle A.16, Kaufmann M.17, Neubauer A.18, Schäfer-Eckart K.19, Hänel M.20, Peceny R.21, Frickhofen N.22, Kiehl M.23, Giagounidis A.24, Görner M.25, Repp R.26, Link H.27, Kiani A.28, Naumann R.29, Brümmendorf T.H.5, Serve H.10, Ehninger G.4, Berdel W.E.3, Müller-Tidow C.1, StudienAllianz Leukämie (SAL) 1

The AML-AZA trial (n = 214 pts) tested whether azacytidine (Aza) in combination with intensive chemotherapy could improve outcome in AML (arm A) compared to patients receiving chemotherapy only (arm B). In the whole study population, event free survival (EFS) and overall survival (OS) did not differ significantly between the two groups. DNMT3A mutations were found in 56 of 167 (34%) patients analysed, predominantly in exon 23 (35/167; 21%). Patients with DNMT3A mutation in exon 23 treated with Aza showed trends to longer EFS (median 8.7 versus 5.6 months, p = 0.069) and OS (median not reached versus 13 months, p = 0.28). Cox-regression analysis was performed to evaluate the interaction between DNMT3A exon 23 mutations and treatment. Presence of DNMT3A mutation was associated with trends for prolonged EFS in the AZA arm (HR = 0.64; 95% CI 0.31–1.31, p = 0.221) but not in the control arm. As a result, the hazard ratio (HR) of Aza versus no Aza was decreased to a factor 0.37 when comparing patients with present versus absent DNMT3A mutation exon 23 (ratio of Hazard Ratios = 0.50/1.36 = 0.37; 95%CI 0.15–0.91, p = 0.03). To complement these findings with mechanistic data, U937 cells were transduced with either DNMT3A-wildtype (WT) or DNMT3A-R882H (MT). U937-MT cells showed reduced growth and colony formation in the presence of azacytidine whereas U937-WT cells were resistant. Very low concentrations of azacytidine in combination with cytarabine were effective in inhibiting colony formation in U937-MT cells. U937-WT cells were less affected. Taken together, these data suggest that DNMT3A-R882 mutations are especially susceptible to azacytdine in combination with chemotherapy. A confirmatory clinical trial is required to validate these findings.

Association of single nucleotide polymorphisms of innate immunity and invasive fungal disease in patients with acute myeloid leukemia Schnetzke U.1, Fischer M.1, Spies-Weißhart B.1, Schrenk K.1, Gruh B.2, Wittig S.2, Glaser A.3, Hochhaus A.1, Scholl S.1 Universitätsklinikum Jena, Klinik für Innere Medizin II, Jena, Germany, Universitätsklinikum Jena, Klinik für Kinder- und Jugendmedizin, Jena, Germany, 3Universitätsklinikum Jena, Institut für Humangenetik und Anthropologie, Jena, Germany 1 2

Introduction: Induction chemotherapy of acute myeloid leukemia (AML) is associated with a high risk of invasive fungal disease (IFD). Several pattern recognition receptors (PRR) of the innate immune system are highly important for the response to fungal infections. A series of functionally relevant single nucleotide polymorphism (SNPs) of such PPRs represent promising candidates to predict the individual risk of IFD for intensively treated AML patients. We sought to investigate the impact of different SNPs of innate immunity on the occurrence of IFD during AML induction chemotherapy. Methods: Genotyping of TLR1 (rs5743611, rs5743618, rs4833095), TLR2 (rs5743708), Dectin-1 (rs7309123, rs3901533, rs16910526) and DCsign (rs4804800, rs11465384, rs7248637) was performed by TaqMan method and/ or pyrosequencing in 186 adult AML patients. IFD was classified according to the EORTC/MSG consensus guidelines. Multiple logistic regression analysis was applied to evaluate the association between distinct SNPs or defined SNP clusters and the occurrence of IFD. Results: Patients carrying either two homozygous TLR1 SNPs (rs5743618, rs4833095), the TLR2 polymorphism, the Dectin-1 SNP rs7309123 or polymorphisms of all analyzed DCsign loci have a significant higher risk for developing pulmonary IFD. Importantly, cluster analyses defined as the detection of polymorphisms in at least two subsets of these SNPs (TLR1, TLR2, Dectin-1 and/ or DCsign) demonstrates positivity in only 6 of 138 AML (4.3%) patients without IFD compared to 14 of 48 (29.2%) patients

Abstracts


CONTENTS AUTHOR INDEX

who fulfilled the criteria of IFD according to the EORTC/MSG guidelines (OR 9.06 (95% CI 3.24–25.32), p < 0.001). Conclusions: Defined SNP clusters of innate immunity may predict an increased risk of IFD in AML patients undergoing induction chemotherapy. Disclosure: Ulf Schnetzke: Expert Testimony: Fa. Gilead Förderprogramm Infektiologie 2013. Sebastian Scholl: Expert Testimony: Fa. Gilead Förderprogramm Infektiologie 2013. V428

Non-myeloablative conditioning (NMA) followed by hematopoietic stem cell transplantation (HCT) in patients (PTS) with acute myeloid leukemia (AML): Prognostic impact of the European LeukemiaNet (ELN) standardized reporting system

Fig. 1. ELN1. Disclosure: No conflict of interest disclosed. V429

Bill M.1, Jentzsch M.1, Schubert K.1, Knyrim M.1, Grimm J.1, Cross M.1, Vucinic V.1, Franke G.N.1, Pönisch W.1, Behre G.1, Lange T.1,2, Niederwieser D.1, Schwind S.1

Minimal residual disease-guided preemptive treatment in patients with AML and MDS – a retrospective survey within the SAL study group

1

Universität Leipzig, Hämatologie und Internistische Onkologie, Leipzig, Germany, 2Asklepios Klinik Weißenfels, Hämatologie und Internistische Onkologie, Weißenfels, Germany

Bulycheva E.1, Mayer J.2, Krüger W.3, Mies A.1,4, Šustkova Z.2, Račil Z.2, Prochazkova J.2, Hirt C.3, Ringhoffer M.5, Berdel W.6, Serve H.7, Röllig C.1, Sockel K.1, Bornhäuser M.1, Ehninger G.1, Thiede C.1,4, Platzbecker U.1,4

Introduction: Introduction of NMA rendered older or unfit pts eligible for HCT. The prognostic impact of the ELN system for these pts is unknown. Patients and methods: We analyzed 215 pts, treated at the University Hospital Leipzig between April 1999 & July 2013 (median age 64 years [y; range 37–76y]). We included de novo (n = 123; 57.2%), secondary (n = 72; 33.5%) or therapy-related (n = 19; 8,8%) AML. Pts received: 2Gy total body irradiation either with fludarabine (30 mg/m² at day -4 to -2; n = 211; 98.1%) or without (n = 4; 1.9%). Donors were HLA-matched related (n = 35; 16.3%), HLA-matched (n = 119; 55.3%) or mismatched (>=1 antigen; n = 61; 28.3%) unrelated. Cytogenetics, presence of FLT3ITD & NPM1 & CEBPA mutation (mut) status at diagnosis were assessed centrally. Results: For all pts the median overall survival (OS) after transplantation was 1.98y, with a 3y-OS of 45.1%. Pts were classified according to the ELN system: 20.5% favorable (fav; n = 44; including 18.2% [n = 8] with core-binding factor AML, 34.1% [n = 15] CEBPA mut & 53.3% [n = 23] NPM1 mut/no FLT3-ITD with NK), 24.2% intermediate-I (int-I; n = 52; including 17.3% [n = 9] NPM1 mut/FLT3-ITD, 13.5% [n = 7], NPM1 wild type [wt]/FLT3-ITD, 67.3% [n = 35] NPM1 wt/no FLT3-ITD with NK), 23.1% intermediate-II (int-II; n = 50; including 2.0% [n = 1] with t(9;11)) & 32.1% adverse (adv; n = 69; including 71.0% [n = 49] with complex karyotype; 31.9% [n = 22] with -7; 40.6% [n = 28] with -5/del5q; 2.9% [n = 2] with inv(3)) genetic risk. We analyzed the cumulative incidence of relapse (CIR) according to ELN groups & found a significant separation (p = 0.033; Figure 1A). This was also found by trend for OS (p = 0.088; Figure1B). Analyzing OS among each group, only fav was performing significantly better than int-II (p = 0.043) or adv (p = 0.010). Conclusion: We analyzed the prognostic impact of the ELN system for AML pts receiving NMA-HCT. Several studies focused on younger (≤60y), chemotherapy-based consolidated pts, showed that fav performed best & adv worst. Int-II had better outcome than int-I. In our set only fav pts performed better than int-II or adv pts. Thus, especially older or unfit pts within the int-I or adv ELN group might benefit from NMA-HCT.

1

Abstracts

Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Medical Department I, Dresden, Germany, 2University Hospital Brno, Department of Internal Medicine – Hematology and Oncology, Brno, Czech Republic, 3Ernst-Moritz-Arndt-Universität Greifswald, Department of Internal Medicine C, Hematology and Oncology, Marrow Transplantation, Greifswald, Germany, 4German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany, 5Städtisches Klinikum Karlsruhe, Department of Internal Medicine III, Karlsruhe, Germany, 6Universitätsklinikum Münster, Department of Medicine A – Hematology, Oncology and Pneumology, Münster, Germany, 7 J. W. Goethe-Universität Frankfurt, Department of Medicine II, Hematology/ Oncology, Frankfurt, Germany

Introduction: Detection of minimal residual disease (MRD) in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) in complete hematological remission (CR) offers a potential therapeutic window allowing early interventions in order to prevent overt hematological relapse. Since prospective clinical studies are limited we initiated a retrospective survey within the SAL study group and members of the DKTK Consortium to document the current clinical use of the approach and clinical outcomes. Patients: We analyzed 74 patients (pts) at four centers (m/f = 42/32; median age 54 yrs (19–74)) with either MDS (n = 7) or AML (n = 67) in CR after either conventional intensive chemotherapy (CTx; n = 18) or allogeneic hematopoietic stem cell transplantation (allo-HSCT; n = 56), undergoing preemptive therapy. Markers for MRD monitoring were mutations, cytogenetic aberrations or donor chimerism for transplanted pts. Twenty-eight pts had a normal karyotype; 13 pts – complex; 6 pts carried -7 and/or inv(3); other aberrations were detected in 25 pts. Among molecular aberrations, the most prominent was NPM1 (n = 17) and FLT3-ITD (n = 10), followed by CBFβ/MYH11 (n = 7), RUNX1/RUNX1T1 (n = 7) and MLL/AF6 (n = 3). Results: The median time to MRD positivity after completion of intensive therapy was 7.5 months (range, 1–97). Subsequent MRD-guided treatment in pts treated only with CTx included additional intensive CTx (13%), hypomethylating agents (HMA, 23%), clofarabine (23%), targeted therapy (gemtuzumab ozogamycin, 10%), low-dose cytarabine (13%), and allo-HSCT (13%). In transplanted patients the most preferred treatment for the molecular relapse was donor lymphocyte infusion (23 pts, 41%; median number of DLI = 2, range, 1–6), alone or in combination (60% with HMA). 26 MRD-based treated pts (38%) did not experience a hematological relapse and were alive (17 pts, median observation time 959 days, range, 297–3647), or died due to another cause (9 pts) with median survival of 359 days (range, 125–954). In 8 pts without hematological relapse more than one MRD recurrence was observed. In the remaining 62% relapsing patients, median time to hematological relapse was 267 days (range, 24–1161) after MRD positivity.

Oncol Res Treat 2015;38(suppl 5):1–270

123


CONTENTS AUTHOR INDEX

Conclusions: The retrospective analysis demonstrates that MRD-guided therapies have already entered routine practice in patients with MDS/ AML. Therefore, clinical trials are needed to better define molecular markers and subsets of patients who might benefit from this approach. Disclosure: Ekaterina Bulycheva: Other Financial Relationships: Gilead. Uwe Platzbecker: Financing of Scientific Research: Celgene, Novartis.

Freier Vortrag

Mammakarzinom I V430

An AKT/SOX2 molecular axis drives breast carcinoma clonogenicity Schaefer T.1, Wang H.1,2, Mir P.2, Pereboom T.1, Merz B.3, Fehm T.4, Perner S.5, Rothfuss O.3, Kanz L.2, Schulze-Osthoff K.3,6, Lengerke C.1,2,7 Department of Biomedicine, University Hospital, Basel, Switzerland, Department of Internal Medicine II, University Hospital, Tuebingen, Germany, Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen, Germany, 4Women’s Hospital, University Hospital, Duesseldorf, Germany, 5 Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital, Bonn, Germany, 6 German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany, 7Clinic for Hematology, University Hospital, Basel, Switzerland 1 2 3

Introduction: The embryonic transcription factor SOX2 (Sex determining region Y – Box 2) plays key roles in embryonic stem cell self-renewal. More recently SOX2 protein was detected in various types of cancer including breast carcinoma (BC). The molecular regulation of SOX2 in BC remains largely unknown. Here, we describe a functional dependence of SOX2 on the Ser/Thr-kinase AKT, a central mediator of cell growth, proliferation and metabolism, whose deregulation is a common determinant in BC. Methods: Eight human BC cell lines and 10 patient samples were analyzed for SOX2, AKT and pAKT expression by RT-PCR, immunoblot, and IHC. Cells were cultured under conventional or 3D-conditions enriching for putative cancer stem cells (CSCs) and treated with cisplatin, paclitaxel or respectively MK-2206, AKT1/2 and AKT siRNAs to inhibit AKT activity. SOX2 expression was modulated with lentiviruses containing shSOX2 or an inducible mCherry-SOX2-overexpression construct. A previously described lentivirus containing a DsRed-SOX2 regulatory region 1 reporter element was used to isolate SOX2-positive breast CSCs. Tumor sphere assays were applied to investigate clonogenicity. pAKT and SOX2 subcellular localization was followed by confocal and life-time microscopy. Results: Both pAKT and SOX2 were induced by culture conditions promoting CSCs and functionally regulated BC cell clonogenicity in tumor sphere assays. Unlike conventional chemotherapies, which promoted the outgrowth of SOX2-positive putative CSCs, AKT-inhibition reduced cell growth without enhancing the frequency of SOX2-reporter positive cells in the surviving fraction. Mechanistically, AKT activity modulated SOX2 subcellular distribution promoting non-canonical localization in the cytosol and subsequent proteasomal degradation. Immunoprecipitation showed direct physical interaction of AKT and SOX2 proteins and permutation of the T116 AKT phosphorylation site fostered cytoplasmic retention of SOX2. Functionally, SOX2 overexpression rescued tumor sphere formation in anti-AKT treated BC cells, indicating SOX2 as a major molecular target mediating AKT-effects on clonogenicity. Conclusions: We here describe a hitherto unrecognized coupling of AKT and SOX2 proteins in human BC that has utmost significance for clonogencity and therapy resistance. Our investigations reveal important mechanistic details about the promises of AKT-inhibitor therapies that are currently under laboratory and clinical investigation in BC.

V431

Mcl-1 confers protection of HER2+ breast cancer cells exposed to hypoxia: Therapeutic implications Bashari M.H.1, Fan F.1, Vallet S.1, Arn M.2, Cardone M.H.2, Beckhove P.3, Schneeweiss A.1, Sattler M.4, Opferman J.T.5, Jäger D.1,6, Podar K.1 National Center for Tumor Diseases (NCT), University of Heidelberg, Medical Oncology, Heidelberg, Germany, 2Eutropics Pharmaceuticals, Inc, Cambridge, United States, 3National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany, 4 Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States, 5 St. Jude Children’s Research Hospital, Memphis, United States, 6German Cancer Research Center (DKFZ), Applied Tumor-Immunity, Heidelberg, Germany 1

Introduction: Breast cancer (BC) is the most common malignancy and the leading cause of cancer-related mortality in women. HER2 overexpression occurs in 15–20% of BC patients and is associated with poor clinical outcome. Hypoxic conditions develop during cancer progression. Molecular mechanisms, which lead to the adaptation of HER2+ BC cells to hypoxia are unknown. The anti-apoptotic Bcl-2 family member Mcl1 is frequently amplified in BC; and elevated Mcl-1 levels correlate with poor prognosis. The aim of this study is to evaluate whether Mcl-1 plays a role in the adaptation of HER2+ BC cells to hypoxic conditions and to provide the derived rationale to therapeutically target Mcl-1. Methods: The molecular role of Mcl-1 in HER2+ BC cells under hypoxic conditions was investigated using genetical as well as pharmacological approaches followed by proliferation, survival, and spheroid forming assays, subcellular fractionation, RT-PCR as well as immunoprecipitation/ blotting. Mcl-1(Δ/null) Murine Embryonic Fibroblasts (MEFs) were used to verify the specific anti-Mcl-1 activity of the novel small molecule inhibitor EU-5346 (EUTROPICS, Cambridge). Results: We demonstrate for the first time a strong correlation between rapid induction of Mcl-1 and HIF-1α under hypoxic conditions in HER2+ BC cells. Genetic depletion of Mcl-1 decreased HER2 and HIF-1α levels followed by inhibition of BC cell survival. Surprisingly, Mcl-1 protein levels did not change after genetic depletion of HER2, indicating a regulatory role of Mcl-1 upstream of HER2. The functional interrelation of Mcl-1 and HER2 under hypoxic conditions was supported by Mcl-1/HER2 co-localization within the mitochondrial fraction as well as the formation of a Mcl-1/HER2- protein complex. Moreover, our data demonstrate that Mcl1 stabilizes HER2 protein levels via inhibition of ubiquitination. Given its proposed key role in the survival of cancer cells in general, Mcl-1 is the current focus of widespread cancer drug development efforts. Indeed, EU-5346 specifically blocks the interaction of Mcl-1 with pro-apoptotic BH3-only proteins. Similar to genetically targeting Mcl-1 EU-5346 induced cell death and decreased spheroid formation in HER2+ BC cells. Conclusions: Taken together, our data demonstrate the critical role of Mcl-1 in HER2+ BC cell survival under hypoxic conditions and provide the preclinical framework for the therapeutic use of novel Mcl-1-targeting agents to improve patient outcome in BC. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

124

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V432

V433

GAIN2: Adjuvant phase III trial comparing an intensified dosedense adjuvant therapy with EnPC compared to a dose-dense, dose-adapted therapy with dtEC-dtD in patients with primary high risk breast cancer: Results of the second safety interim analysis

Routine care reality of patients with metastatic breast cancer 1995–2014: Improvement of survival is restricted to hormoneand HER2-receptor-positive tumors

Möbus V.1, Lück H.-J.2, Forstbauer H.3, Wachsmann G.4, Ober A.5, Schneeweiss A.6, Christensen B.7, Ekkehard E.8, Grischke E.-M.9, Höffkes H.-G.10, Klare P.11, Ko Y.D.12, Schmatloch S.13, Burchardi N.14, Loibl S.15, von Minckwitz G.14, German Breast Group

1

Klinikum Frankfurt Höchst GmbH, Frankfurt am Main, Germany, Gynäkologisch-Onkologische Praxis am Pelikanplatz, Hannover, Germany, 3 Hämatologisch-Onkologische Schwerpunktpraxis, Troisdorf, Germany, 4 Klinikum Sindelfingen-Böblingen, Böblingen, Germany, 5St. Vincenz Krankenhaus, Limburg, Germany, 6Universitäts-Klinikum, Heidelberg, Germany, 7 Ruppiner Kliniken GmbH, Neuruppin, Germany, 8Klinikum, Mutlangen, Germany, 9Universitätsklinikum, Tübingen, Germany, 10Klinikum, Fulda, Germany, 11Praxisklinik Krebsheilkunde und Brustzentrum, Berlin, Germany, 12 Tumorzentrum, Bonn, Germany, 13Elisabeth-Krankenhaus, Kassel, Germany, 14 German Breast Group, Neu-Isenburg, Germany, 15German Breast Group, GBG Forschungs GmbH, Neu-Isenburg, Germany 1 2

Introduction: Combined chemotherapy requires compromises in terms of dosage and treatment interval due to toxicities. Sequential administration of monotherapies allows high doses of single substances and dose-dense intervals. Such regimens proved to be very effective in early breast cancer with high risk of recurrence. Nab-paclitaxel (nP) leads to a more favorable toxicity profile and greater efficacy compared with solvent-based taxanes. Methods: The GAIN2 study compares toxicity and efficacy of a pre-defined intense dose-dense regimen (EnPC; E:150 mg/m2, nP:330 mg/m², C:2000 mg/m²) with a dose-dense regimen, where single doses are adjusted depending on individual hematological and non-hematological toxicities (dtEC-dtD; E:38–120 mg/m², C:450–1200 mg/m², D: 60–100 mg/m²). Primary endpoint is the invasive disease-free survival in patients with primary node-positive or high-risk node-negative breast cancer (luminal A: ≥LN; luminal B: N+; HER2pos/TNBC: N0/N+) A first safety interim analysis after 200 with completed chemotherapy has been published (Noeding et al. Ann Oncol 2014). The second safety interim analysis with 900 treated patients will be presented. Results: In terms of hematological adverse events, the rates of febrile neutropenia grade 3–4 (12% vs. 8%) and thrombopenia grade 3–4 (12% vs. 5%) was significantly higher in the EnPC arm. As for the non-hematological side effects grade 1–4, the rate of increased alkaline phosphatase (59% vs. 40%), increased ALAT (69% vs. 59%), peripheral neuropathy (83% vs. 68%), arthralgia (63% vs. 49%), myalgia (48% vs. 41%), and bone pain (25% vs. 17%) was significantly higher in the EnPC arm whereas the rate of epistaxis (10% vs. 25%), edema (13% vs. 26%), and hand-footsyndrome (12% vs. 28%) was significantly higher in the dtEC-dtD arm. There were no differences between the treatment arms for the toxicities of special interest (cranial nerves affections, macula degenerations, anaphylactic reactions). In the EnPC arm, 30% required dose-reductions due to hematological toxicities compared to only 10% in the dtEC-dtD arm (p < 0.001). The dose could be escalated to the maximum in more than a third of the patients receiving dtEC-dtD. In 9% of women a reduction was required in the 4th cycle of docetaxel. Conclusion: Due to similar toxicity profiles, the study will be continued without changes. Disclosure: No conflict of interest disclosed.

Weide R.1, Feiten S.2, Friesenhahn V.2, Heymanns J.1, Kleboth K.2, Thomalla J.1, van Roye C.1, Köppler H.1 Praxisklinik für Hämatologie und Onkologie, Koblenz, Germany, 2Institut für Versorgungsforschung in der Onkologie, Koblenz, Germany

Introduction: Improvements in survival rates of patients suffering from metastatic breast cancer have been found in randomized clinical trials over the past years. Due to necessary selection criteria it is difficult to transfer these results into routine care. The aim of this analysis was to determine overall survival of unselected patients who received their treatment in an oncology group practice. Methods: Retrospective analysis of all patients with metastatic breast cancer who were seen between 06/1995 and 12/2014. Relevant clinical data were transferred from clinical files into a database and analyzed statistically using SPSS and SURVSOFT. Results: Data of 767 female patients with a median age of 61 years (31–93) (time of metastasis) were analyzed. 81% were postmenopausal, 23% suffered from primary metastatic disease. Sites of metastases were distributed as follows: 49% visceral, 34% bone, 9% lymph nodes, 4% CNS and 4% others. Mean number of metastatic sites was 1.5 (1–6). 77% of tumors were hormone-receptor-positive, 18% HER2-positive and 9% triple-negative. Median overall survival was 35 months (0–321+), disease specific survival was 37.3 months. Disease specific 5-year survival rate was 34%. Overall survival was significantly correlated with the sites of metastases, number of involved organs, disease-free survival since initial diagnosis and hormone-receptor status. Patients with a hormone-receptor-positive tumor had a median overall survival of 38 months (0–321+) compared to patients with a triple-negative tumor, who showed a median overall survival of 13 months (0–109+). Patients with a HER2-positive tumor had a median overall survival of 39 months (0–189+). Comparisons of disease specific survival with registry data from the US, Great Britain and different German areas revealed a significant better survival after 1 year (86% versus 44–73%) and after 5 years (34% versus 13–25%). A trend, although statistically non-significant, showing an continuous improvement in survival rates could be observed. Patients who received diagnosis between 1995 and 1999 had a median disease specific survival of 30.5 months. Patients who were diagnosed 2010 or later lived 43.0 months in comparison. Conclusions: Survival rates of patients suffering from metastatic breast cancer with hormone-receptor- and / or HER2-positive tumors can be extended significantly in routine care. This is probably due to the sequential use of targeted therapies. Disclosure: No conflict of interest disclosed. V434

EVI1 promotes breast cancer cell growth via MAPK signaling, but independent of the estrogen Wang H.1,2, Schäfer T.1, Konantz M.1, Reich S.2, Braun M.3, Perner S.3, Varga Z.4, Moch H.4, Kanz L.2, Schulze-Osthoff K.5,6, Lengerke C.1,2,7 Department of Biomedicine, University Hospital, Basel, Switzerland, Department of Internal Medicine II, University Hospital, Tuebingen, Germany, 3 Department of Prostate Cancer Research, Institute of Pathology, University Hospital of Bonn, Bonn, Germany, 4Institute of Surgical Pathology, University Hospital, Zurich, Switzerland, 5Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen, Germany, 6German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany, 7Clinic for Hematology, University Hospital, Basel, Switzerland 1 2

Introduction: The transcriptional regulator EVI1 has been mainly studied in myeloid leukemia, but expression has also been detected in solid tumors. While almost no data exist on EVI1 in breast carcinoma (BC), a previous report suggests that overexpression may indicate poor clinical

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

125


CONTENTS AUTHOR INDEX

outcome in estrogen receptor (ER) negative tumors (Patel et al., 2011). Here, we analyze the roles and molecular targets of EVI1 in human BC. Methods: EVI1 expression was analyzed by qRT-PCR and immunoblot analysis in 8 human BC cell lines and a tissue microarray (TMA) containing 373 patient BC samples surveyed by immunohistochemistry. Lentiviruses containing EVI1-shRNAs were used to generate EVI1-knockdown cells, which were then compared to control-treated cells in various assay formats: cell cycle, BrdU, apoptosis (PI and Annexin-V/7-AAD staining, Caspase 3/7 and PARP activity), immunophenotypic, and qRT-PCR. Xenotransplantation experiments were performed in zebrafish embryos as described (Konantz et al., 2012). Smad3 siRNAs were introduced by transient transfection and β-estradiol (50–100 nM) used for cell culture and zebrafish treatment. Results: EVI1 was expressed in most breast cancer samples independent of ER status. EVI1 gene expression levels influenced long-term survival in ER- (p = 0.026) but not ER+ patients. In BC cell lines and primary cells, EVI1 knockdown impaired growth by suppressing proliferation and enhancing apoptosis sensitivity. The growth reduction was observed in both EVI1-knockdown ER- as well as ER+ cells, but in the latter β-estradiol could provide a functional rescue. Consistently, in vivo addition of β-estradiol could rescue tumor initiation and growth in zebrafish transplanted with ER+ but not ER- BC cells. On the molecular level, knockdown of EVI1 led to a cell cycle arrest by de-repressing its known target gene SMAD3 and thereby suppressing the cell cycle proteins p21 and p15. siRNA against SMAD3 only partially rescued growth in EVI1-knockdown cells, indicating that additional pathways are also involved. Indeed, EVI1 knockdown strongly inhibited the MAPK-pathway, thereby additionally regulating BC cell proliferation. Conclusion and discussion: EVI1 is widely expressed amongst breast carcinoma cells where it influences apoptosis and proliferation via SMAD3 repression and activation of the MAPK-pathway. The precise molecular mechanisms underlying the effect of EVI1 on MAPK activity are currently under investigation. Disclosure: No conflict of interest disclosed. V435

Cascade of toxicological tests: breast cancer treatment affects trophoblast cells in 2D and explant culture Fröhlich K.1, Schmidt A.1, Heger J.1, Lupp A.2, Avemarg S.1, Markert U.R.1 Friedrich Schiller University, Placenta Laboratories, Department of Obstetrics, Jena, Germany, 2Friedrich Schiller University, Institute of Pharmacology and Toxicology, Jena, Germany 1

Introduction: Pregnancy and breast cancer is a rare coincidence, but breast cancer is one of the most common malignancies during pregnancy. Guidelines on treatment for breast cancer during pregnancy already exist and demonstrate that pregnancy-associated breast cancer (PABC) can be treated according to recommendations for non-pregnant women with breast cancer. Nonetheless, a decreased birth weight is often observed in newborns which may be due to harmful effects of chemotherapy on trophoblast cells. Therefore, we aimed to determine the toxicity of chemotherapeutic drugs on these cells. Methods: HTR-8/SVneo and JEG-3 cells cultured as monolayers were grown in 96-well plates. Placental villous tissue explants (PVTE) were obtained after spontaneous delivery or cesarean section (n = 3). Trophoblast cells and PVTE were incubated with doxorubicin, docetaxel, 5-fluorouracil or vincristine for at least 48 hours. The metabolic activity was evaluated via MTS assay. Supernatant analyses of glucose, lactate, lactate dehydrogenase (LDH), human chorionic gonadotropin (hCG), estrogen and progesterone were performed. Data were analyzed with SPSS 22 using a linear mixed model. Furthermore, PVTE were embedded in paraffin for histological examinations. Results: The metabolic activity of PVTE, as evaluated via MTS assay, was only reduced by doxorubicin or docetaxel, but not by 5-fluorouracil or vincristine treatment. In contrast, the metabolic activity of HTR-8/SVneo and JEG-3 cells was significantly reduced by all tested drugs. Glucose, lac-

126

Oncol Res Treat 2015;38(suppl 5):1–270

tate and LDH as potential markers of toxicity were significantly affected in the supernatant of PVTE treated with the different chemotherapeutics. In contrast, expression of hCG, estrogen and progesterone was not modified. Hematoxylin/eosin staining revealed more morphological anomalies in 5-fluorouracil or vincristine than in doxorubicin or docetaxel treated PVTE. Conclusion: Toxicological investigations on trophoblast cell lines enable the detection of harmful effects of chemotherapy but fail to simulate in vivo conditions. Therefore, PVTE display a suitable tool for studying effects of different chemotherapeutic drugs during pregnancy. Among the analyzed parameters, only the metabolic markers glucose, lactate and LDH and the MTS assay appear to be useful for detecting harmful effects and are recommended for further investigations. Hematoxylin/eosin staining indicates early toxic morphological changes, even when the MTS assay did not. Disclosure: No conflict of interest disclosed.

Posterdiskussion

Allogene Transplantation P436

Reduced platelet transfusions and earlier platelet engraftment using alemtuzumab based conditioning regimen in allogeneic SCT Neumann T.1, Schneidewind L.2, Thiele T.3, Schulze M.1, Klenner A.1, Busemann C.1, Pink D.4, Greinacher A.3, Dölken G.1, Krüger W.1 Universitätsmedizin Greifswald, Klinik für Innere Medizin C – Hämatologie / Onkologie, Greifswald, Germany, 2Universitätsmedizin Greifswald, Klinik für Urologie, Greifswald, Germany, 3Universitätsmedizin Greifswald, Institut für Immunologie und Transfusionsmedizin, Greifswald, Germany, 4HELIOS Klinikum Bad Saarow, Klinik für Hämatologie, Onkologie und Palliativmedizin, Bad Saarow, Germany 1

In patients undergoing allogeneic stem-cell transplantation (SCT) conditioning regimens containing alemtuzumab instead of antithymocyte globulin (ATG) may result in an earlier platelet engraftment and a reduced number of platelet transfusions. We performed a retrospective, single center, case-control study analyzing time to engraftment and transfusion needs using alemtuzumab in comparison to ATG as part of conditioning protocol. Median values for time to platelet engraftment, number of transfused platelet concentrates and number of transfused red cell concentrates were 12 vs. 19.5 days (p < 0.001), 2 vs. 14 (p < 0.001) and 6 vs. 14.5 (p = 0.003) in the alemtuzumab and ATG group. Time to leukocyte engraftment did not differ significantly with median 15 days in both groups. Patients in the ATG group showed significant lower median platelet counts at the day of stem-cell infusion (38 vs. 95.5 Gpt/l, p = 0.008) and higher values for median C-reactive protein (CRP) after first antibody infusion (69.0 vs. 43.6 mg/l, p = 0.001) compared to alemtuzumab group. Test for significance was done by using Wilcoxon rank sum test. Subgroup analysis considering the type of ATG used (Thymoglobulin vs. ATG Fresenius) revealed that differences between alemtuzumab and ATG group were more due to effects of ATG Fresenius than Thymoglobulin. Median values for time to platelet engraftment, number of transfused platelet concentrates and number of transfused red cell concentrates were 14 vs. 23 days, 5 vs. 21 and 8 vs. 21 in the Thymoglobulin and ATG Fresenius group. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

P437

CD19+CD21low B-cells as biomarker for the prediction and monitoring of response to immunosuppressive therapy for chronic graft-versus-host disease Kralj M.1, Kuzmina Z.1, Weigl R.1, Rottal A.2, Körmöczi U.2, Pickl W.2, Greinix H.3 Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria, 2Institute of Immunology, Medical University of Vienna, Vienna, Austria, 3 Department of Internal Medicine, Medical University of Graz, Graz, Austria 1

Chronic graft-versus-host disease (cGVHD) is a serious, life-threatening complication of allogeneic hematopoietic stem cell transplantation (HCT). Afflicted patients present heterogeneous organ manifestations varying in severity and patients’ impairment of quality of life. Here, we investigated cellular biomarkers for the prediction of clinical response to various immunosuppressive agents (ECP, cyclosporine A (CsA), tacrolimus and sirolimus) in the course of cGVHD. Different B-cell subsets, inluding CD19+CD21low B-cells were analyzed by flow cytometry in the peripheral blood (PB) of cGVHD patients at the onset of immunosuppressive therapy (IS) and again 6 months after start of therapy. At the same time patients were evaluated for the presence of clinical signs and symptoms of cGVHD according to NIH recommendations. Different B-cell subsets were compared overall between responders and non-responders to IS and within each IS treatment group. The study included 144 patients with a median age of 39 (range, 17–70) years. Response to therapy was defined as decrease in severity for at least 1 score and no new organ manifestations of cGVHD. Overall the response rate to IS at 6 months after start of treatment was 54% (78 out of 144 patients). 47 patients received ECP, 51 CsA, 18 tacrolimus and 28 sirolimus. The overall absolute numbers of CD19+ B-cells in PB at the start of IS therapy were significantly lower in non-responders compared to responders (157 × 106/L vs 307 × 106/L, p = 0.003). Overall, relative numbers of CD19+CD21low B-cells measured at the onset of IS were significantly elevated in patients that later did not respond to therapy compared to the ones that did (17.94% vs 13.55%, p = 0.017). A similar trend was observed within the sirolimus and tacrolimus IS groups. At 6 months after start of IS, percentages of CD19+CD21low B-cells were significantly higher in non-responders compared to responders (20.16% vs 7.55%, p < 0.001). Similar significant observations were seen in all four therapy groups. By comparing relative values of CD19+CD21low B-cells at the onset of IS and 6 months after, we observed a significant decrease in patients who responded to therapy (13.55% vs 7.55%, p < 0.001). In patients who did not respond to IS, these values stayed elevated (17.94% vs 20.16%, p = 0.243). Our results show that CD19+CD21low B-cells could serve both as predictive biomarker for response to cGVHD treatment as well as for monitoring disease activity in cGVHD. Disclosure: No conflict of interest disclosed. P438

Cytomegalovirus induce apoptosis and alloresponse in acute leukemia cells Koldehoff M.1, Lindemann M.2, Opalka B.3, Mühlenberg T.4, Ross R.S.5, Elmaagacli A.H.1,6 University of Duisburg-Essen, Department of Bone Marrow Transplantation, Essen, Germany, 2University of Duisburg-Essen, Institute for Transfusion Medicine, Essen, Germany, 3University of Duisburg-Essen, Department of Hematology, Essen, Germany, 4University of Duisburg-Essen, Department of Medical Oncology, Essen, Germany, 5University of Duisburg-Essen, Institute of Virology, Essen, Germany, 6HELIOS Schwerin, Department of Oncology and Hematology, Schwerin, Germany 1

Introduction: Cytomegalovirus (HCMV) reactivation occurs frequently after hematopoietic stem cell transplantation and is associated with an increased treatment-related mortality. Induction of apoptosis by HCMV is unusual because HCMV utilizes various strategies to prevent apoptosis in infected cells in order to delay cell death and maintain viral replication.

Abstracts

Cells and Methods: Various acute leukemic cell lines were purchased for in vitro experiments, and further bone marrow or peripheral blood stem cell samples were collected from healthy volunteers after informed consent in accordance with institutional guidelines. For infection we used the AD169 HCMV strain (ATCC-VR-538, American Type Culture Collection, Manassas, VA, USA). Various different cellular and molecular biological studies with HCMV-infected cells were performed. Results: Here we show that HCMV can infect the acute leukemia cell lines Kasumi-1 (AML) and SD-1 (BCR-ABL-positive ALL), which inhibited their proliferation and induced apoptosis in almost all cells after 14 days. Although HCMV induced a significant up-regulation of the anti-apoptotic gene cFLIP and the anti-stress gene Gadd45a, and simultaneously down-regulated the pro-apoptotic genes p53, Gadd45gamma in Kasumi-1 and SD-1 cells, we found that these anti-apoptotic mechanisms failed in HCMV-infected acute leukemia cells and apoptosis occurred via a caspase-dependent pathway. ELISpot data indicated that peripheral blood mononuclear cells (PBMC) of healthy individuals had significantly higher numbers of interferon-gamma, granzyme B and perforin secreting cells when stimulated with Kasumi-1 with vs. without HCMV infection; indicating that alloresponses were enhanced due to HCMV infection of the Kasumi-1 cell line. In addition, HLA-DR expression as determined by FACS analysis was significantly increased on PBMC of healthy individuals when stimulated with Kasumi-1 with vs. without HCMV infection. Finally, proliferation of Kasumi-1 with vs. without HCMV infection was significantly lower. Conclusions: We conclude that HCMV can provide anti-leukemic effects in vitro. To determine if this phenomenon may be clinically relevant further investigations will be required. Disclosure: No conflict of interest disclosed. P439

No impact of geographic distance to patient residence on the outcome of allogeneic hematopoietic cell transplantation (alloHCT): Retrospective analysis in a JACIE-accreditated transplant center Brand S.1, Dietrich S.2, Hegenbart U.2, Bondong A.2, Luft T.2, Schönland S.2, Ho A.D.2, Görner M.1, Dreger P.2 Städtische Kliniken, Bielefeld, Germany, 2Universitätsklinikum Heidelberg, Innere Medizin V, Heidelberg, Germany 1

The purpose of this study was to assess the impact of distance to residence on alloHCT outcomes in a JACIE-accreditated center in Germany. Study design and patients: In a single center retrospective analysis, the distance to primary residence was assessed for all consecutive patients who underwent alloHCT for hematological malignancy 2005 and 2011 and assigned to 5 categories: Local (0–50 km), regional A (>50–100 km), regional B (>100–150 km), national (>150 km), and international. The impact of distance category on non-relapse mortality (NRM), relapse, progression-free survival (PFS), and overall survival (OS) was studied by log-rank comparisons. Results: A total of 562 patients were assigned to local (229), regional A (166), regional B (76), national (75), and international residence (15). Median age was 53 (18–70) years; diagnoses were myeloid malignancies (53%), lymphoma (16%), myeloma (13%), CLL (10%), and ALL (8%); disease status was refractory at alloHCT in 17%; median EBMT score (Gratwohl) was 2 (0–7). Donors were matched siblings (31%), unrelated donors (68%), and other (1%). Compared to local/regional patients, the national/international group contained significantly less patients with myeloid malignancy (40% vs 56%, p < 0.0001), significantly more patients with myeloma (25% vs. 10%), and had a significantly poorer EBMT score (3 vs 2, p < 0.0001), but was matched for all other baseline variables. There were no differences between local, regional A and regional B patients for any survival endpoint. Similarly, NRM (HR 1.04; 95% CI 0.58–1.87) and OS (HR 0.85; 95%CI 0.61–1.2) were not significantly different between pooled local/regional patients and national/international patients. However, local/regional patients had a significantly lower relapse risk (HR

Oncol Res Treat 2015;38(suppl 5):1–270

127


CONTENTS AUTHOR INDEX

0.59; 95% CI 0.4–0.89; p 0.011), translating to a significant PFS advantage (HR 0.69; 95% CI 0.49–0-97; p 0.032). This effect disappeared after adjusting for diagnosis. Significant predictors of PFS were disease status (refractory unfavorable), EBMT score, and diagnosis (myeloma unfavorable, CLL favorable). Conclusion: This study failed to detect a detrimental impact of distance from residence to transplant center on NRM and disease control after alloHCT for hematological malignancies. Albeit further studies are needed, these results suggest that distance to HCT center should not overrule criteria such as JACIE accreditation and centre experience when selecting a transplant center for a specific patient. Disclosure: No conflict of interest disclosed. P440

GTPase Rap1 regulates the shear stress-resistant adhesion of Mesenchymal Stromal Cells (MSCs) Steinmann J.1, Fleck E.2, Henschler R.2, Fisch P.1, Scheele J.1, Kropshofer H.3 University Hospital Medical Center Freiburg, Department of Pathology, Freiburg, Germany, 2Institute for Transfusion Medicine and Immune Hematology, German Red Cross Blood Service, Medical Center of the Johann Wolfgang Goethe University, Frankfurt, Germany, 3F. Hoffmann- La Roche Ltd., Pharmaceuticals Division, Basel, Switzerland 1

Introduction: Mesenchymal stromal cells (MSCs) are increasingly used as an intravenously applied cellular therapy. Rap1 is a major regulator of integrin activation, and therefore a candidate molecule to modulate homing properties of MSCs. We here studied the adhesion of human and murine MSCs to endothelial cells and recombinant integrin ligands and their homing behavior in mice. Methods: MSCs deficient in Rap1A and/ or Rap1B, mediated via siRNA medicated knockdown or derived from the respective knockout mice were investigated with regards to MSC migration and adhesion in a flow-chamber model. Results: siRNA mediated knockdown of Rap1A or Rap1B led to decrease in both basal and Stromal Derived Factor-1 (SDF-1) induced firm adhesion of MSCs in a flow-chamber model, whereas knockdown of both Rap1A+B also inhibited arrest of flowing MSCs to inflammatory endothelial cells. Further siRNA experiments indicated that Rap1-Guanosin Nucleotide Exchange Factor (GEF) PDZ-GEF1 is responsible for constitutive adhesion of MSCs to Vascular Cell Adhesion Molecule (VCAM)-1, whereas treatment of MSCs with siRNA against EPAC2 inhibited predominantly the adhesion induced by the chemokine, SDF-1. We next established Rap1 deficient MSCs from Rap1A knockout mice. Whereas homozygous MSCs were not viable in culture, heterozygous Rap1A +/- MSCs recapitulated the adhesion deficit observed in human MSCs. The EPAC stimulator 8-Br-(4-PCT)-cAMP induced Rap1 activation and increased adhesion of MSCs. Homing experiments in tumor bearing mice indicated a shift in the homing pattern of Rap1A deficient mMSCs compared with wt MSCs, away from lung, liver and hematopoietic tissues towards muscle, kidney and lymph nodes. Conclusions: Together, our findings demonstrate an essential role for Rap1mediated pathways in MSC migration, and provide a strategy to engineer MSC grafts in an effort to improve their therapeutic efficiency. Disclosure: No conflict of interest disclosed.

P441

Invasive mycosis after allogeneic stem cell transplantation at the university medical centre of Würzburg – Incidence of the disease as well as morbidity and mortality of patients with the condition Möller A.-K.1, Einsele H.2, Heinz W.J.2 Klinikum Heilbronn GmbH, Innere Medizin I, Heilbronn, Germany, Universitätsklinikum Würzburg; II. Medizinische Klinik, Würzburg, Germany

1 2

Introduction: Invasive fungal infections (IFI) still present an important and life threatening complication especially for recipients of an allogeneic stem cell transplantation (SCT). Despite improved diagnostics, new prophylactic options and therapies, mortality and morbidity of patients with a systemic mycosis remain at a high level. Knowledge about the incidence, timing and impact of IFI is relevant to choose the appropriate diagnostic and antifungal strategy. Methods: To record the incidence of IFI among patients after allogeneic SCT, a prospective, non-interventional study was conducted. A primary endpoint was to analyse the morbidity and mortality rates of patients with IFI. The clinical course of the patients was actively monitored until 100 days after the start of the conditioning chemotherapy and a follow-up examination was carried out after six months. IFI were classified according to the EORTC-MSG criteria revised in 2008. Results: Within six months a total of 41 patients were included in the survey. During this study 22% of the patients contracted a probable and 17% a possible invasive mycosis. One probable mycosis was caused by Fusarium spp., the remaining infections were ascribed to Aspergillus spp. (94%). At the end of the first month the risk to develop a probable mycosis was at 17%, which increased to 22% at the end of the sixth month. The patients without evidence of an IFI were hospitalized for SCT for an average of 41.8 days. Patients with IFI spent an average of 55.9 days at the hospital, revealing a significant difference in the length of hospital stay (p = 0.015). Deducting two possible cases with unknown survival, in the first half year after SCT 16% of patients without IFI had passed away while 20% of those with a possible aspergillosis and 66.6% of those with a probable mycosis had died. The estimated cumulative survival time of the patients without systemic mycosis was 170.9 days and of those with IFI was 131.8 days (p = 0.016). The death of the patients with a possible or probable infection occurred an average of 40 days after the initial diagnosis of the IFI. Conclusion: In the survey presented here, a high incidence of invasive IFI after SCT was detected. The mortality of the affected patients was significantly higher than in SCT recipients without IFI. Improved diagnostic options may increase the rate of diagnosed infections. Prophylactic and therapeutic strategies have to be adapted to the clinical situation. Disclosure: No conflict of interest disclosed. P442

Allogeneic stem-cell transplantation for early relapse of multiple myeloma after autologous stem-cell transplantation Schmidt V.1, Mügge L.-O.1, Klink A.1, Hilgendorf I.1, Hochhaus A.1, Sayer H.G.1,2 Klinik für Innere Medizin II, Universitätsklinikum Jena, Hämatologie und internistische Onkologie, Jena, Germany, 24. Medizinische Klinik, HELIOSKlinikum Erfurt, Hämatologie und internistische Onkologie, Hämostaseologie, Erfurt, Germany 1

Introduction: Prognosis of patients with early relapse of multiple myeloma (MM) after conditioning with melphalan followed by autologous hematopoietic stem-cell transplantation (autoHSCT) is poor, and treatment options are limited. Methods: We report a retrospective analysis of 12 MM patients (8male/4female; median age 49 (37–63) years) who received an allogeneic HSCT (alloHSCT) between 03/2008 and 09/2014 at our center. Before alloHSCT, patients had received a median of 5 (range 2–6) therapy lines including a median of 2 (1–3) previous autoHSCTs. Relapse occurred 7 (3–9) months after the last autoHSCT or 12 (12–18) months in 3 patients who received

128

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

maintenance therapy with bortezomib. Conditioning therapy consisted of treosulfan/fludarabin (9/12), melphalan/fludarabin (2/12) or fludarabin/ TBI (1/12). HSCT was performed from a matched related donor (1/12), from an HLA-matched (10/10) unrelated donor (7/12), or a mismatched unrelated donor (4/12). ATG was added in case of unrelated donor HSCT. Results: After alloHSCT, 2 patients (17%) achieved a complete remission, 3 patients (25%) reached a very good partial remission, and 6 patients (50%) a partial remission. One patient (8%) experienced a graft failure, and despite an autologuous rescue transplantation died due to relapse of MM within 4 months. After a median follow up of 55 months, 7 patients (58%) are still alive and median overall survival has not been reached yet. A relapse occurred in 8 patients (66%). Median progression free survival was 8 months. An acute GvHD (≥ grade III) occurred in 5 patients, but was sensitive to GvHD-directed therapy. Three patients (25%) who developed a mild to moderate chronic GvHD are still alive and in a stable remission 42, 50, and 69 months after alloHSCT, even though they received no further MM-directed therapy and were not in a CR early after alloHSCT. All patients (5/12) with a deletion of chromosome 13 (del13q) together with a translocation involving chromosome 14 [t(4;14) or t(11;14)] died within 31 (median 13) months after alloHSCT. In comparison to the other 7 patients, the difference was significant at p < 0.01 (Fisher exact test). Conclusion: AlloHSCT is a feasible therapeutic option for heavily pretreated patients with an early relapse of MM after autoHSCT. Especially patients with chronic GvHD may profit from a graft-versus-myeloma effect. The combination of del13q and either t(4;14) or t(11;14) is a strong negative predictor, even after alloHSCT. Disclosure: No conflict of interest disclosed. P443

Steroid-induced hyperglycemia adversely impacts outcome in graft-versus-host disease Stauber M.1, Aberer F.2, Zebisch A.1, Neumeister P.1, Greinix H.T.1, Sill H.1, Sourij H.2, Wölfler A.1 Medizinische Universität Graz, Klin. Abteilung für Hämatologie, Graz, Austria, Medizinische Universität Graz, Klin. Abteilung für Endokrinologie und Stoffwechsel, Graz, Austria 1 2

Introduction: Graft-versus-host disease (GvHD) is a common complication after allogeneic stem cell transplantation. Since the current first-line treatment is based on high-dose glucocorticoid therapy, steroid-induced hyperglycemia develops frequently in these patients. However, little is known about the impact of hyperglycemia on outcome in GvHD. Methods: We performed a retrospective analysis of 100 patients, who received systemic glucocorticoid therapy due to acute or chronic GvHD and investigated the impact of hyperglycemia on outcome in these patients. For each subject regular blood glucose measurements during glucocorticoid therapy and transplant- as well as GvHD-related parameters, such as underlying disease, time to onset of GvHD after transplantation, number of affected organs, response to glucocorticoids, insulin therapy and (fungal) infectious complications were recorded. For analysis of the impact of hyperglycemia, the median glucose level was taken and patients were divided into 4 groups (median glucose level < 100 mg/dl, 100–120 mg/dl, 121–150 mg/dl and >150 mg/dl). Results: With a median of 42 blood glucose measurements per patient, only nine patients had a median glucose level <100 mg/dl and were therefore assigned to group 1. In 29 cases the median glucose value was between 101 and 120 mg/dl (group 2), 30 patients were assigned to group 3 and the remaining 32 cases displayed a median glucose level >150 mg/dl. While median overall survival (OS) was not reached in group 1, increasing median glucose levels were associated with decreasing median OS of 20, 17 and 8 months for groups 2, 3 and 4, respectively (p < 0,01). Accordingly, 5-year OS was significantly lower for group 4 when compared to all other groups (p < 0,01) and for group 2 and 3 when compared to group 1 (p < 0,01). In addition, insulin treatment, which was given in 49 patients, was associated with a shorter median OS (8 versus 29 months, p < 0,01). While the number of relapses was low (n = 9) and not affected by hyper-

Abstracts

glycemia, non-relapse related mortality was responsible for most cases of death (n = 51). In particular, patients with invasive fungal infections had a poor outcome (median OS of 5 months versus 24 months, p < 0,001). Conclusions: In this retrospective analysis, we observed an adverse impact of glucocorticoid-induced hyperglycemia on outcome in patients with GvHD. Prospective trials testing a stringent glycemic control are therefore urgently needed in this clinical setting. Disclosure: No conflict of interest disclosed. P444

Minimal toxicity conditioning with fludarabine and 2 Gy total body irradiation for allogeneic hematopoietic cell transplantation Dörfel D.1,2, Mück F.1, Vogel W.1, Wirths S.1, Möhle R.1, Kanz L.1, Faul C.1, Bethge W.A.1 Universitätsklinik Tübingen, Innere Medizin II – Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Tübingen, Germany 1

Introduction: Non-myeloablative conditioning with fludarabine (FLU, 3×20 mg/m2) and 2 Gy total body irradiation (TBI) is currently the least toxic conditioning regimen used for the treatment of hematologic malignancies with hematopoietic cell transplantation (HCT) primarily in an elderly or comorbid patient population. Methods: We report retrospectively our single center experience with this regimen in 40 consecutive patients undergoing allogeneic HCT after minimal toxicity conditioning with FLU/2Gy TBI followed by postgrafting immunosuppression using a calcineurininhibitor combined with mycophenolate mofetil with or without sirolimus from 2004 to 2014. Results: 40 adult patients received allogeneic HCT during this period for the treatment of acute myeloid leukemia (n = 25), non-Hodgkin lymphoma (n = 10), myelodysplastic syndrome (n = 4), and multiple myeloma (n = 1). The median age at time of HCT was 62 (range 25 to 76) years. Peripheral blood stem cells were exclusively used as graft source. Donors were matched related (n = 5), matched unrelated (n = 33) or mismatched unrelated (n = 2). Median follow-up was 13 months with a Kaplan Meier estimated overall survival (OS) for all patients of 55%, 37% and 32% at 1, 3, and 5 years, respectively. There was no significant difference in OS/ EFS in different age groups. The non-relapse mortality (NRM) at day 100, 1 year and 3 years was 5%, 20% and 23%. 38% of patients developed a relapse of the underlying disease (AML 10/25 = 40%, NHL 3/10 = 30%, MDS 1/4 = 25%, MM 1/1 = 100%). The cumulative incidence of acute GvHD was 23% (≥II° 13%) and of chronic GvHD was 73% (limited = 43%, extensive = 30%). Conclusion: In conclusion, our data suggest that RIC with FLU/TBI enables long-term survival even in an elderly and comorbid patient population. However, incidence of chronic GVHD is high, which may have a negative impact on quality of life. Disclosure: No conflict of interest disclosed. P445

Urological complications and BK virus associated hemorrhagic cystitis under allogenic stem cell transplantation Schneidewind L.1, Neumann T.2, Burchardt M.1, Krüger W.2 Universitätsmedizin Greifswald, Klinik für Urologie, Greifswald, Germany, Universitätsmedizin Greifswald, Klinik für Innere Medizin C, Hämatologie/ Onkologie, Greifswald, Germany 1 2

Introduction: Every year 50.000 patients receive a stem cell transplantation worldwide, but there is a lack of data focussed on urological complications under this therapy. To fill this gap of knowledge we performed a retrospective analysis of all adult patients undergoing allogenic stem cell transplantation from 01/2011 to 06/2013 in our institution.

Oncol Res Treat 2015;38(suppl 5):1–270

129


CONTENTS AUTHOR INDEX

Methods: All patient records of adult patients with hematological diseases undergoing their first allogenic stem cell transplantation from 01/2011 to 06/2013 were collected. The statistical analyses was performed with SPSS 22.0. Statistical tests performed were Pearson´s correlation, Qui-Square testing and logistic regression. Results: We identified 39 patients (22 males and 17 females). Twenty five patients (64.1%) had a urological complication. Most frequent complications were bacterial urinary tract infection (n = 13; 33.3%), acute renal failure (n = 6; 15.4%) and BK virus associated hemorrhagic cystitis (n = 5; 12.8%). We observed an association of creatinine increase (about 20 µmol/l at time of onset of BK viruria) with BK viruria (Pearson`s correlation 0.64; p = 0.01) and BK viruria is significantly linked to acute renal failure (Pearson´s correlation 0.35; p = 0.029). In univariate regression BK viruria is significant linked to urological complication (p = 0.025). We could not identify a significant risk factor for urological complications in multivariate analysis. Conclusion: The most frequent urologic complications in adult allogenic stem cell transplantation are bacterial urinary tract infection and BK virus associated hemorrhagic cystitis. We suggest that BK virus infection during stem cell transplantation can also lead to BK virus associated nephropathy. Disclosure: No conflict of interest disclosed. P446

Significance of serum galactomannan for diagnosing invasive fungal disease in patients after allogeneic hematopoietic stem cell transplantation Rieger C.1, Peterson L.1, Lustig D.1, Fiegl M.1, Ostermann H.1 Ludwig-Maximilians-Universität München, Medizinische Klinik III, München, Germany 1

Objectives: Immunosuppressed patients frequently develop infections with Aspergillus spp., leading to severe illness or death. Early diagnosis and prompt initiation of antimycotic treatment is the key to success in fighting invasive fungal infections. Seropositivity for Galactomannan plays a significant role for diagnosing as well as the ensuing therapy of invasive fungal infection in everyday clinical practice. We were wondering if the engraftment status in recipients of hematopoetic stem cell grafts has an impact on the serum level of Galactomannan. Methods: Galactomannan, C-reactive protein and the course of leukocyte regeneration were recorded after studying medical charts. Inclusion criteria were allogeneic hematopoietic stem cell transplantation (HSCT) in the years 2011 to 2013 as well as at least one positive Galactomannan in the observational period (days –25 until +25 before or after engraftment). Results: 52 patients (w: 24 m: 28) with a median age of 50 years (max.: 67 years, min.: 20 years) and a median length of stay of 62 days (max. 249 days, min: 36 days) in hospital for HSCT (10 bone marrow transplants; 43 PBSCTs) were recorded. Underlying diseases were AML (26 patients/49,1%), ALL (7pts./13,2%), MM (5pts./8,6%), CML (4 patients/6,9%), Lymphoma (3pts./5,7%), OMF (3pts./5,2%), MDS (2pts./3,4%), CLL (1pt./1,9%), vSAA (1pt./1,7%) and acute biphenotypic leukemia (1pt./1,7%). Conclusion: Having noticed peaking of serum-Galactomannan in multiple patients around engraftment, preliminary data indicate that leukocyte recovery might indeed impact on the serum level of Galactomannan in recipients of hematopoetic stem cell grafts. Our data suggest that this might reflect true immunologic activity. However, should further studies confirm our data, it will have important implications for the way that Galactomannan is applied as a diagnostic tool for invasive fungal infection in the allogeneic stem cell transplantation.

Posterdiskussion CML P447

Center size, patient age, and co-medication do not impact molecular response to 1st-line nilotinib treatment in chronic myelogenous leukemia (CML): An analysis of the German patients enrolled into ENEST1st Hammersen J.1, Berger C.1, Eigendorff E.1, Schenk T.1, Fabisch C.1, Hochhaus A.1, La Rosée P.1 Universitätsklinikum Jena, Klinik für Innere Medizin 2, Hämatologie/Onkologie, Jena, Germany 1

Introduction: Nilotinib induces rapid, deep, and sustained molecular remission in chronic phase CML. In order to assess efficacy and tolerability post approval (“real life situation”), the European ENEST1st-trial (EudraCT 2009–017775–19) evaluated the rate of molecular response in newly diagnosed CML patients (pts). The German subgroup of ENEST1st was analysed for potential factors influencing treatment results such as center size, age, comorbidities, and relevant co-medication. Patients/Methods: 275 pts were recruited from 06/2010 until 11/2011. Pts with newly diagnosed CML were treated with 300 mg nilotinib twice daily. The primary endpoint of the trial BCR-ABL ≤0.01% according to international scale (IS) or MR4 after 18 months (mo) of treatment was used to test the impact of age, comorbidity, co-medication with focus on cytochrome P450 3A4 (CYP3A4) interacting agents, and center size. Results: 258 of 275 pts (60% male) were evaluable. Median age was 52 years (ys, range, 18–83), ECOG was 0 in 76%. Median follow up was 24 mo. Standardized quantitative RT-PCR for BCR-ABL at 18 mo was evaluable for 73% pts. 85% achieved major molecular response (BCR-ABLIS ≤ 0.1%, MMR), 41% MR4. Females did better with a MR4-rate of 56% vs 32% (p < 0.001). Age did not impact MR4-rate, when patients were divided into subgroups <65 vs ≥65 ys (40% vs 47%, n.s.). Of note, pts younger than 30 ys (n = 25) had a lower rate of MR4 vs pts older than 30 ys (21% vs 44%, p = 0.03). Pts older than 60 ys received a median of 8 drugs as co-medication, pts younger than 60 ys had a median of 5 drugs (p = 0.01). A detailed analysis of comorbidities will be given. 19% of all patients received CYP3A4-interacting substances, and showed an increased rate of MR4 (56% vs 38%, p = 0.05). No significance was shown for CYP3A4 inductors and inhibitors. 39% pts were treated in study centers with < 5 pts, 17% in centers with ≥10 pts. Center size conferred no significant impact on MMR. In large centers, the rate of missed 3-monthly PCR analyses was significantly lower (7% vs 20%, p = 0.04). Conclusion: High efficacy of nilotinib in newly diagnosed CML was confirmed. No significant impact on outcome was shown for higher age, co-medication and center size. These data are in line with similar analyses from study groups (i.e. GHSG, Cologne) suggesting that inclusion of pts in trial protocols safeguards high quality of care, and allows systematic evaluation of variables potentially affecting the quality of care. Disclosure: Jakob Hammersen: No conflict of interest disclosed. Paul La Rosée: Financing of Scientific Research: Received speakers honorarium from Novartis; Other Financial Relationships: Received travel support from Novartis.

Disclosure: No conflict of interest disclosed.

130

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

P448

Hydroxyurea (HU) supresses the growth of CML cells expressing BCR/ABLT315I in heavily pretreated patients and synergizes with ponatinib in inhibiting the proliferation of CML cells in vitro Schneeweiss M.1, Herndlhofer S.1, Byrgazov K.2, Lion T.2, Sperr W.R.1, Valent P.1,3, Gleixner K.1 Medizinische Universität Wien / Universitätsklinik für Innere Medizin I, Abteilung für Hämatologie und Hämostaseologie, Wien, Austria, 2Children‘s Cancer Research Institute, Wien, Austria, 3Medizinische Universität Wien / Ludwig Boltzmann Cluster Oncology, Wien, Austria 1

Introduction: In chronic myeloid leukemia (CML), the occurrence of BCR/ABLT315I leads to resistance against most tyrosine kinase inhibitors (TKI) and is therefore a major clinical challenge. Ponatinib, which supresses BCR/ABLT315I, cannot be used in all patients because of severe side effects. Other therapeutic strategies, such as drug combinations or bridging to allogenic stem cell transplantation (aSCT), are often considered in these patients. Hydroxyurea (HU) is a drug that interferes with DNA-synthesis and has been used for the treatment of CML since decades. Patients and methods: In this study, the follow-up of 4 CML-patients (all men, aged 39–65 years, all in chronic phase) treated with HU (1–3 g/day) was analysed. In all patients, BCR/ABLT315I was detected after treatment with two or three TKI. Expression of total BCR/ABL and BCR/ABLT315I was analyzed by qPCR at several time points. To evaluate the effect of the combination “HU + ponatinib” on cell proliferation in vitro, K562 and KU812 cells were incubated with HU, ponatinib, or the combination of both drugs before 3H-thymidine uptake was measured. Results: In all 4 patients treated with HU, leukocyte counts and total BCR/ ABL levels remained stable for a time period of 3–12 months. Surprisingly, in 3 of 4 patients, the leukemic subclone expressing BCR/ABLT315I was no longer detectable during and after HU-treatment. In one patient, the percentage of BCR/ABLT315I compared to total BCR/ABL sunk significantly from 94% to 7% during HU therapy as assessed by mutation-specific ligase-dependent qPCR. After 3 months, 2 of 4 patients were transplanted. In the 2 other patients who were not eligible for SCT, the disease remained stable for 6 and 12 months, respectively. We next questioned whether HU would cooperate with ponatinib in suppressing leukemic cell growth. As assessed by 3H-thymidine uptake experiments, HU and ponatinib, used in suboptimal concentrations, were found to synergize in inhibiting the proliferation of K562 and KU812 cells. This was confirmed by Calcusyn-software. Conclusion: Together, we show that HU suppresses the growth of CML clones harbouring BCR/ABLT315I. This observation is of particular interest for heavily pretreated patients, especially when bridging to SCT is required. Furthermore, we show that HU synergizes with ponatinib in inhibiting the proliferation of CML cells in vitro. Whether this drug combination is also effective in vivo remains to be evaluated in further studies. Disclosure: No conflict of interest disclosed. P449

Marked regression of myelofibrosis during reduced-dose dasatinib treatment in chronic myelogenous leukemia in accelerated phase Schelker R.C.1, Huber E.1, Herr W.1, Vogelhuber M.1 Uniklinikum Regensburg, Innere Medizin III (Hämatologie/Onkologie), Regensburg, Germany 1

Introduction: Severe myelofibrosis (MF) occurs in 10% of patients with chronic myelogenous leukemia (CML) at the time of diagnosis and is associated with poor prognosis. For this subgroup of patients, allogeneic haematopoietic stem cell transplantation (HSCT) should be considered from the start of tyrosine kinase inhibitor (TKI) treatment given the risk for depletion of the bone marrow (BM) hematopoietic stem cell pool. One major possibility to avoid HSCT could be TKI induced reduction of MF.

Abstracts

Authors have described significant regression of MF during treatment with imatinib but data regarding other TKIs are very sparse. Methods: One patient with CML in accelerated phase (AP) was treated after informed consent first with imatinib 400 mg/day p.o., after developing severe myalgia with dasatinib 100 mg/day p.o. and after neutropenic fever episode with dasatinib 80 mg/day p.o.. Treatment response was frequently monitored and remission was defined according to standard criteria. BM biopsy was done in the University Hospital of Regensburg (Germany) after informed consent. To reduce the risk of only fokal diagnostic events, follow-up biopsies were done alternating on the right-left side of the pelvis. BM cellularity was evaluated by hematoxylin and eosin stain; reticulin and collagen abundance and intersection potential by gomori silver method. Results: We describe here a patient with CML in AP, who developed marked regression of MF from MF-3, the highest MF grade according to European Myelofibrosis Network (EUMNET) Grading, to MF-0 (normal BM) with focally residual fibrosis (MF-1) following 2 years and 4 months of reduced-dose dasatinib (80 mg daily) treatment with very good haematologic, cytogenetic and molecular response. As far as we are aware, this is the first report proving marked MF reduction during dasatinib treatment. Discussion: In general two main reasons for MF reduction during dasatinib treatment need to be discussed. One option is that the first event concerns remission of CML by TKI therapy, leading secondary to regression of MF (indirect effect). The second possibility is that independent of BCR-ABL inhibition dasatinib is able to cause MF reduction directly. Potential mechanisms by which dasatinib impacted MF in this patient could be: inhibition of BCR-ABL which can uncouple canonical JAK2/ STAT5 pathway; suppression of Lyn (member of Src family kinase) fused with ETV6; interference with PDGFRß; decrease in dysplastic and small megakaryocytes. Disclosure: No conflict of interest disclosed. P450

Cost-effectiveness analysis of ponatinib in the treatment of CML in Germany le Coutre P.1, Hirt C.2, Iannazzo S.3, Brenn J.4, Chiroli S.5 Charité Universitätsmedizin, Berlin, Germany, 2Greifswald University Medical Center, Greifswald, Germany, 3SIHS, Torino, Italy, 4ARIAD Pharmaceuticals (Germany) GmbH, Frankfurt, Germany, 5ARIAD Pharmaceuticals, Epalinges, Switzerland 1

Introduction: In chronic-phase chronic myeloid leukemia (CP-CML), highly-resistant patients (e.g. third line or beyond) have a very poor prognosis. Current preferred treatment options are tyrosine kinase inhibitors (TKIs) and allogeneic stem cell transplantation (allo-SCT) for suitable patients. There are few therapeutic options for patients who exhibit resistance or intolerance to first- and second-generation (2G) TKIs (nilotinib, dasatinib, or bosutinib) or who have the T315I mutation. Ponatinib was designed to inhibit the kinase activity of native BCR-ABL and all mutant variants, including T315I. Ponatinib efficacy in patients with highly-resistant CML was demonstrated in the pivotal phase II Ponatinib Ph+ ALL and CML Evaluation (PACE) trial. In the absence of head-to-head trials, an economic model employing a German public healthcare perspective was developed to assess the cost-effectiveness of ponatinib for the treatment of patients with CP-CML compared to current third-line treatment options in Germany. Methods: The cost-effectiveness model compares ponatinib, 2G TKIs, and allo-SCT, with cost per life-years (LY) saved and cost per quality-adjusted life-years (QALYs) gained as outcome measures, and a lifetime time horizon. Resource use includes study drugs, monitoring and follow-up, adverse events and allo-SCT procedure. Costs, based on current tariffs in Germany, and benefits (LY/QALYs) were discounted at 3.5% per annum. To address uncertainty, we performed sensitivity analyses (SA) to identify parameters that most strongly influenced results. Clinical validity was evaluated by comparing model-generated survival estimates with relevant clinical data.

Oncol Res Treat 2015;38(suppl 5):1–270

131


CONTENTS AUTHOR INDEX

Results: Over the patients’ lifetime, ponatinib offers an increase in LY of almost 6 years and a gain of almost 4 QALYs compared with the nextbest therapy. When only time in the CP-CML disease stage is considered, the use of ponatinib results in a substantial increase in time-in-state. The incremental cost-effectiveness ratios range from € 6,486/QALY gained vs. allo-SCT to € 26,574/QALY gained vs. bosutinib. SA showed the model was robust to plausible changes in input parameters and had good face validity. Conclusions: This analysis suggests that treating CP-CML with ponatinib provides a substantial clinical benefit as compared with current alternatives at a reasonable cost, from the perspective of the German public healthcare system. Sensitivity analysis confirmed the robustness of these results. Disclosure: Philipp le Coutre: Advisory Role: ARIAD Pharmaceuticals, Inc., Novartis, BMS, Pfizer: Advisory Board member; Financing of Scientific Research: ARIAD Pharmaceuticals, Inc., Novartis, BMS, Pfizer: Honoraria. Silvia Chiroli: Employment or Leadership Position: ARIAD Pharmaceuticals employee; Stock Ownership: ARIAD Pharmaceuticals,Stock owner. P451

Cytogenetic and molecular responses in German patients with chronic-phase chronic myeloid leukemia (CP-CML) in a prospective observational study (SIMPLICITY) Repp R.1, Hehlmann R.2, Linde H.3, Reiser M.4, Tesch H.5, Foreman A.6, Schreiner L.7, Hillig G.7 Sozialstiftung Bamberg, Hämatologie und internistische Onkologie, Palliativmedizin, Bamberg, Germany, 2Universität Heidelberg, Mannheim, Germany, 3MVZ für Blut-und Krebserkrankungen, Potsdam, Germany, 4Praxis Internistischer Onkologie und Hämatologie, Köln, Germany, 5Onkologische Gemeinschaftspraxis am Bethanien-Krankenhaus, Frankfurt, Germany, 6ICON Plc, San Francisco, United States, 7Bristol-Myers Squibb, München, Germany

Fig. 1.

Conclusions: The German cohort of SIMPLICITY showed trends similar to the total study population, with greater proportion of pts on 2nd generation TKIs achieving CCyR and MMR and reaching ‘optimal’ or ‘warning’ status, and fewer in the ‘failure’ category compared with pts on a 1st generation TKI. Disclosure: Roland Repp Grit Hillig: Employment or Leadership Position: Employed by Bristol-Myers Squibb.

1

Introduction: Observational studies can provide information about prescribing patterns and clinical practice not available from randomised trials. SIMPLICITY (NCT01244750) is an ongoing, global observational study of CP-CML patients (pts) receiving first-line (1L) treatment with a tyrosine kinase inhibitor (TKI) outside of clinical trials. This analysis presents clinical response data for SIMPLICITY pts in German centres. Methods: Pts in SIMPLICITY receive imatinib (IM), dasatinib (DAS) or nilotinib (NIL) in academic centres and community practices in the US and Europe. Clinical response following start of 1L TKI therapy was assessed by cytogenetic response (CyR) (karyotype or FISH) and molecular response (MR) (PCR on the international scale). Descriptive statistics are presented. ‘Optimal’, ‘warning’ or ‘failure’ responses are defined by ELN 2013 guidelines. Results: 1207 pts (European: 35%) were enrolled prospectively through 22Sept2014, receiving IM (n = 415), DAS (n = 416) or NIL (n = 376). Of these, 151 were enrolled at German centres (13% of the total study population) and received IM (n = 46), DAS (n = 55) or NIL (n = 50). Of 115 German pts followed for 12 mos from start of 1L TKI, 35% (n = 40/115) were tested for CyR and 18% (n = 8/45; 13 pts tested), 49% (n = 17/35; 17 pts tested) and 29% (n = 10/35; 10 pts tested) achieved complete CyR (CCyR) for IM, DAS, and NIL cohorts respectively. Among tested pts 88% (n = 35/40) achieved CCyR. By 12 mos, 73% (n = 84/115) of German pts followed had been tested for MR and 27% (n = 12/45; 29 pts tested), 54% (n = 19/35; 29 pts tested) and 40% (n = 14/35; 26 pts tested) achieved major MR (MMR), for IM, DAS, and NIL cohorts respectively. Among tested pts 54% (n = 45/84) achieved MMR. By 12 mos, the proportion of pts with ‘optimal’, ‘warning’ and ‘failure’ responses varied by 1L TKI (Figure), differing from the total population.

132

Oncol Res Treat 2015;38(suppl 5):1–270

P452

Efficacy and safety of nilotinib in routine healthcare: Treatment of CML patients (pts) failing prior therapy – Results of the non-interventional TARGET study Dengler J.1, le Coutre P.2, Müller M.C.3, Stegelmann F.4, Ulshöfer T.5, Sauer A.6, Reichert D.7, Schwinger U.8, Waller C.F.9, Schardt C.10, Losem C.11, Schneider-Kappus W.12, Stern S.13, Vehling-Kaiser U.14, Meincke M.15, Frank O.15, Ottmann O.G.16 Onkologische Schwerpunktpraxis, Heilbronn, Germany, 2Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany, 3 Medizinische Fakultät Mannheim der Ruprecht-Karls-Universität Heidelberg, III. Medizinische Klinik, Mannheim, Germany, 4Universitätsklinikum Ulm, Innere Medizin III, Ulm, Germany, 5Schwerpunktpraxis für Hämatologie und Onkologie, Ludwigsburg, Germany, 6Medizinisches Mehrversorgungszentrum für Blut- und Krebserkrankungen, Potsdam, Germany, 7Onkologische Gemeinschaftspraxis, Westerstede, Germany, 8Internistische Schwerpunktpraxis Onkologie, Hämatologie, Gastroenterologie, Stuttgart, Germany, 9Universitätsklinikum Freiburg, Innere Medizin I, Freiburg, Germany, 10Onkologische Praxis und Tagesklinik, Gelsenkirchen, Germany, 11Schwerpunktpraxis für Hämatologie und Onkologie, Neuss, Germany, 12Praxis für Hämatologie und Onkologie, Ulm, Germany, 13Praxisklinik für integrative Onkologie, Altötting, Germany, 14 Tagesklinik, Landshut, Germany, 15Novartis Pharma GmbH, Nürnberg, Germany, 16Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt a.M., Germany 1

Introduction: Nilotinib (NI) as a potent and highly selective BCR-ABL tyrosine kinase inhibitor is indicated for Ph+ CML pts in CP and AP with intolerance to or resistance of prior therapy including imatinib (IM) as well as for de novo Ph+ CML in CP. Methods: Follow-up analysis of an observational study of NI in 449 pts with Ph+ CML resistant to or intolerant of prior treatment (2nd line or higher) within routine clinical management in 149 centres in Germany (Jan 2008-April 2015). Results: 59.2% of the pts were older than 60 yrs and 8.0% older than 80 yrs (median age 65.1 yrs). 99.1% (0.7%) presented in CP (AP)(phase missing in 1pt). 92% had a good performance status (Karnofsky index ≤1). 93.1% of all pts were pretreated with IM (any dose). Further prior drug treatments were chemotherapy (23.6%), IFN (16.7%), dasatinib (18.9%) and other unspecified drugs (12.7%). Three pts had received SCT in the past (multiple answers possible). 57.8% were treated with NI 2nd line. 48.1%/39.6% were treated with NI mostly due to resistance/intolerance

Abstracts


CONTENTS AUTHOR INDEX

against IM and 4.5%/12.5% against dasatinib, respectively. At initial visit, a dose of 800 mg NI/d was prescribed in 53.2% and 400 mg/d in 17.1%. Median duration of NI was 573 days at the data cut-off for this analysis. Remission status at study entry was 61.7% in CHR, 42.6% in MCyR/26.3% in CCyR (missing data in 19.8%), 30.1%/10.5% in MMR/CMR. These responses improved significantly under NI, reaching cumulative incidences of CHR, MMR and CMR of 90%, 62.6% and 34.3%, respectively. Of note, cytogenetic response improved as well but is not conclusive enough due to missing examinations (e.g. 87.3% after 12 months). Dose reduction or therapy interruption at any time occurred in 29.4% or 15.8%. 75.9% experienced at least one AE during the observation period which was considered serious in 14.7%. 42.6% of pts with documented final visit (n = 357) prematurely discontinued the study. Hematologic AEs were observed in 13.1% of pts, non-hematologic AEs occurred in 38.5% of pts. The most frequently reported AEs (AEs ≥5% are listed) were skin reactions (pruritus 11.8%, rash 10%, alopecia 6.7%), fatigue (10.2%), thrombocytopenia (6.7%), arthralgia (5.6%), gastrointestinal symptoms such as nausea (6.7%) and upper abdominal pain (5.4%) as well as headache (10.2%). Conclusions: In this broad population of CML pts with poor response or intolerance to a prior therapy including IM these data support the use of NI as an efficacious and safe drug. Disclosure: Jolanta Dengler: Employment or Leadership Position: Selbständige Ärztin; Advisory Role: /; Stock Ownership: /; Honoraria: /; Financing of Scientific Research: Vortragshonorare Firma Novartis; Expert Testimony: /; Other Financial Relationships: /; Immaterial Conflict of Interests: / O Ottmann: Advisory Role: Novartis, BMS, Pfizer, Ariad, SUN Pharma, Fusion Pharma; Financing of Scientific Research: Novartis, BMS, Ariad, SUN Pharma; Expert Testimony: Novartis, BMS, SUN Pharma, Fusion Pharma.

script quantification. Current focus on molecular response favors second generation TKIs which have been demonstrated to be of superior efficacy in inducing deep molecular response the majority of patients. However, data on efficacy beyond clinical trials are still rare. Methods: The Austrian CML registry is an ethics committee approved database under the patronage of the ASHO. It is focused on diagnostic parameters e.g. conventional cytogenetics and/or FISH, BCR-ABL transcript levels, blood counts, general laboratory parameters, CML phase, concomitant diseases, CML specific treatment and including stem cell transplantation and adverse effects. Results: We summarize the CML registry data as of April 2015 and focus on treatment response. The CML registry cohort comprised a total of 444 patients who were followed over a median period of 3.5 years. The proportion of older patients (70+ years) increased and at the last follow-up exceeds a quarter of all patients (27%). Most patients are treated with TKIs and firstline TKI therapy is dominated by imatinib. Use of the second generation TKIs nilotinib or dasatinib increases slowly. About three quarter of patients stay on imatinib (296/393) and -closely linked to this pattern of TKI choice- experience in imatinib use exceeds by far treatment duration with any other TKI. The step-wise introduction of international scale compatible assays over time precludes an overall retrospective response analysis. Aiming for an alternative approach we calculated BCR-ABLhalving time per center but differences between the single centers were large.

P453

Tyrosin kinase inhibitors usage and response analysis in the Austrian CML registry cohort Schmidt S.1,2, Wölfler A.3, Greil R.4, Burgstaller S.5, Sliwa T.6, Petzer A.7, Lang A.8, Weltermann A.9, Voskova D.10, Mitterer M.11, Valent P.12, Eberhard N.13, Walder A.14, Geissler K.15, Andel J.16, Häusler C.17, Ludescher C.18, Oexle H.19, Korger M.20, Schnallinger M.21, Schreieck S.22, Krippl P.23, Wiesholzer M.24, Wöll E.25, Geissler D.26, Rochau U.2,27, Siebert U.2,27, Thaler J.5, Gastl G.1 Medizinische Universität Innsbruck, Universitätsklinik für Innere Medizin V – Hämatologie & Onkologie, Innsbruck, Austria, 2ONCOTYROL – Center for Personalized Cancer Medicine, Innsbruck, Austria, 3Medizinische Universität Graz, Klinische Abteilung für Hämatologie I, Graz, Austria, 4Paracelsus Medizinische Privatuniversität, Universitätklinik für Innere Medizin III, Salzburg, Austria, 5Klinikum Wels-Grieskirchen, Abteilung für Innere Medizin IV, Wels, Austria, 6Hanuschkrankenhaus, Medizinische Abteilung – Hämatologie & Onkologie, Wien, Austria, 7Krankenhaus der Barmherzigen Schwestern, Interne I – Onkologie, Hämatologie & Gastroenterologie, Linz, Austria, 8Landeskrankenhaus Feldkirch, Interne E, Feldkirch, Austria, 9aö. Krankenhaus Elisabethinen Linz, Interne I – Hämatologie & Onkologie, Linz, Austria, 10Allgemeines Krankenhaus Linz, Interne III – Onkologie, Linz, Austria, 11Krankenhaus Meran, Innere Medizin, Meran, Italy, 12AKH Wien, Universitätsklinik für Innere Medizin I – Klinische Abteilung für Hämatologie und Hämostaseologie I, Wien, Austria, 13LKH Leoben, Department für HämatoOnkologie, Leoben, Austria, 14aö. BKH Lienz, Internistische Hämatologie und Internistische Onkologie, Lienz, Austria, 15Krankenhaus Hietzing, 5. Medizinische Abteilung, Wien, Austria, 16LKH Steyr, Innere Medizin I, Steyr, Austria, 17Krankenhaus der Stadt Dornbirn, Innere Medizin, Dornbirn, Austria, 18 Ambulatorium für Hämatologie und Onkologie, Innsbruck, Austria, 19LKH Hall, Interne Abteilung, Hall i. T., Austria, 20Barmherzige Brüder Krankenhaus Eisenstadt, Innere Medizin, Eisenstadt, Austria, 21aö. BKH St. Johann i.T., Innere Medizin, St. Johann i.T., Austria, 22BKH Reutte, Innere Medizin, Reutte, Austria, 23LKH Fürstenfeld, Abteilung für Innere Medizin, Fürstenfeld, Austria, 24 Landesklinikum, St. Pölten, Austria, 25Krankenhaus St. Vinzenz Zams, Innere Medizin, Zams, Austria, 26Klinikum Klagenfurt, Medizinische Abteilung, Klagenfurt, Austria, 27UMIT, Public Health, medical decision making, HTA, Hall i. T., Austria 1

Fig. 1. BCR-ABL halving time.

Conclusions: CML first line treatment in Austria is mainly imatinib based. Changes in BCR-ABL quantification assays and ELN-recommendations preclude an overall retrospective response analysis and using BCR-ABL decline proved impractical as well. Multicenter retrospective response analysis will require assay-based rahter than center-based molecular response evaluation. Disclosure: Stefan Schmidt: Financing of Scientific Research: Novartis, BMS, Pfizer, Ariad; Expert Testimony: Novartis Günther Gastl: No conflict of interest disclosed. P454

Measurement of separase proteolytic activity in single living cells by a fluorogenic flow cytometry assay Haaß W.1, Kleiner H.1, Ruppenthal S.1, Müller M.C.1, Hofmann W.-K.1, Fabarius A.1, Seifarth W.1 IIII. Med. Universitätsklinik, Hämatologie und Onkologie, Medizinische Fakultät Mannheim der Universität Heidelberg, Wissenschaftliches Labor, Mannheim, Germany 1

Introduction: CML management according to ELN recommendations is based on the kinetics of tyrosine kinase inhibitors (TKI) response being determined by international scale compatible assays for BCR-ABL tran-

Introduction: ESPL1/Separase, an endopeptidase, is required for centrosome duplication and separation of sister-chromatides in anaphase of mitosis. Overexpression and deregulated proteolytic activity of Separase as frequently observed in human cancers is associated with the occurrence

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

133


CONTENTS AUTHOR INDEX

of supernumerary centrosomes, chromosomal missegregation and aneuploidy. Increased Separase proteolytic activity in a small subpopulation of tumor cells may serve as driver of tumor heterogeneity and clonal evolution in chronic myeloid leukemia (CML). Currently, there is no quantitative assay to measure Separase activity levels in single living cells. Methods: We have designed a flow cytometry-based assay that utilizes a Cy5- and rhodamine 110 (Rh110)-biconjugated Rad21 cleavage site peptide ([Cy5-DREIMR]2-Rh110) as smart probe and intracellular substrate for detection of Separase proteolytic activity in living cells. For assay establishment Separase activity was analyzed in human leukemic cell lines U937, MEG01 and BV173 and in ficollized peripheral blood mononuclear cells from CML patients and healthy donors. For assay validation cell cycle analysis and Western blot immunostaining experiments were performed. Results: As measured by Cy5 fluorescence the cellular uptake of the fluorogenic peptide was fast and reached saturation after 210 min of incubation in U937 cells. Separase activity was recorded as Rh110 fluorescence providing a linear signal gain within a 90–180 min time slot. Compared to a conventional cell extract-based method flow cytometry delivered equivalent results but was more reliable. Furthermore, the problem of vague loading controls and unspecific proteolysis associated with whole cell extracts were bypassed. The flow cytometric Separase assay allows generation of Separase activity profiles that tell us about the number of Separase positive cells within a sample and about the range of intercellular variation in Separase activity levels within a cell population. The assay was successfully used to quantify Separase proteolytic activity in leukemic cell lines and clinical samples. Conclusion: We have established a novel flow cytometry-based Separase activity assay that allows detection and relative quantification of Separase proteolytic activity in single living cells. Using our standardized protocol the FACS-based assay is a highly sensitive, specific, fast and user-friendly tool that allows generation of tissue- and tumor-specific Separase activity profiles. Disclosure: No conflict of interest disclosed. P455

Retrospective analysis of imatinib first and second line in patients with CML in a real world setting and comparison to results of the IRIS and the GIMEMA studies Kiefer-Trendelenburg T.1,2, Gudzuhn A.2, Rexa B.2, Junghanß C.3, Grobe N.4, Hähling D.5, Meyer B.6, Ollech-Chwoyka J.7, Schmidt C.2, Schüler F.8, Dölken G.2, Hirt C.2 Klinik am See, Rüdersdorf, Germany, 2Universitätsklinikum Greifswald, Hämatologie/Onkologie, Greifswald, Germany, 3Universitätsklinikum Rostock, Hämatologie/Onkologie, Rostock, Germany, 4Diakonie Klinikum Dietrich Bonhoeffer, Neubrandenburg, Germany, 5Hämatologisch-Onkologische Praxis, Schwerin, Germany, 6Onkologische Praxis, Greifswald, Germany, 7 Spitaeler Hochrhein GmbH, Waldshut-Tiengen, Germany, 8DRK Krankenhaus Luckenwalde, Hämatologie/Onkologie/Gastroenterologie/Diabetologie, Luckenwalde, Germany 1

Introduction: Imatinib changed the treatment of CML over the last years. The IRIS delivered the available data about patients treated first line with imatinib, the GIMEMA Working Party on CML published data about imatinib as second line treatment. Since results from trials are sometimes not fully comparable with data obtained in a real-world setting, we report the results of imatinib first and second line treatment in CML patients from Mecklenburg-Vorpommern. Patients were treated by oncological university centers, local hospitals and private-practice oncologists. Methods: Using qRT-PCR for BCR-ABL mRNA on the international scale we are performing molecular monitoring for patients with CML. Almost all patients are inhabitants of an area of Germany called Mecklenburg-Vorpommern (23.000 square kilometers). Retrospective analysis of a total of 88 adult patients with CML is reported here. Analysis of 55 patients receiving imatinib first line were compared to results of IRIS. 33 patients treated with imatinib second line were compared to results of the GIMEMA study.

134

Oncol Res Treat 2015;38(suppl 5):1–270

Results: Table 1 shows the results of our retrospective analysis of patients with CML and imatinib first or second line in a real-world setting and the corresponding data of the IRIS and the GIMEMA study. Tab. 1.

1st line IM

2nd line IM

IRIS*

GIMEMA study**

Number of patients

55

33

476

277

Age at time of diagnosis (median)

51

55

48

52

Age at time of starting IM (median)

51

63

48

55

Median follow-up after starting IM in months, (range)

33 (12–89)

62 (2–110)

77

72

MMR after 12/24/60 months (%)

38/60/78

6/15/52

39/–/65– 90***

–/–/ 54–68***

MR4 (%), median until MR4 (months)

47, 18

24, 62

Not reported

Not reported

EFS after 72 months (%)

97

83

88

OAS after 72 months (%)

97

94

89 (60 months)

91

*(Hughes et al., 2010); **(Palandri et al., 2008); ***Calculated based on ­patients in CCyR (complete cytogenetic response).

Conclusions: Our analysis confirms results of clinical trials of first and second line imatinib treatment showing better molecular responses (rates of MMR and MR4) in first line imatinib. Even though there is no statistical difference in EFS and OS, which is most likely due to the low patient number and relative short median follow-up, the molecular response is earlier and deeper in patients treated with first line imatinib. In addition, this study demonstrates that molecular responses in CML patients from a rural area in Germany treated outside of clinical trials are comparable to results of large trials like IRIS or the GIMEMA study. Disclosure: Thomas Kiefer-Trendelenburg: Other Financial Relationships: Novartis. Carsten Hirt: Financing of Scientific Research: Novartis; Expert Testimony: Novartis; Other Financial Relationships: Novartis, Roche. P456

Results of the 3rd interim analysis of the non-interventional MOMENT II-study for determining efficacy and safety of Nilotinib in newly diagnosed Ph+ CML patients in chronic phase Lathan B.1, Sauer A.2, Ulshöfer T.3, Lange E.4, Tebbe S.5, Schulze M.6, Janssen J.7, Meincke M.8, Frank O.8, Tesch H.9 Gemeinschaftspraxis für Hämatologie und Onkologie, Dortmund, Germany, Medizinisches Mehrversorgungszentrum für Blut- und Krebserkrankungen, Potsdam, Germany, 3Schwerpunktpraxis für Hämatologie und Onkologie, Ludwigsburg, Germany, 4Klinik für Hämatologie, Onkologie und Palliativmedizin, Hamm, Germany, 5Onkologische Praxis, Kassel, Germany, 6 Praxis und Tagesklinik für Hämatologie/Onkologie und Palliativmedizin, Zittau, Germany, 7Gemeinschaftspraxis für Hämatologie und Onkologie, Westerstede, Germany, 8Novartis Pharma GmbH, Nürnberg, Germany, 9Onkologische Gemeinschaftspraxis, Frankfurt am Main, Germany 1 2

Introduction: Nilotinib (NI) is a potent and highly selective BCR-ABL TKI approved for treatment of newly diagnosed Ph+ CML pts in CP based on ENESTnd data showing improved treatment with higher molecular response rates vs. imatinib (IM). NI is also indicated for CP and AP Ph+ CML pts who failed prior therapy including IM.

Abstracts


CONTENTS AUTHOR INDEX

Methods: 3rd interim analysis of a non-interventional study of NI in 279 pts with de novo Ph+ CML in CP within routine clinical management (Aug 2011- Mar 2015; 110 centres in Germany). Results: The median age was 57.5 yrs (range 17–88). 43.4% and 2.5% of the pts were older than 60 and 80 yrs. The median time since diagnosis of CML was 12 days (range 0–66). 92.8% of the pts had a good performance status (ECOG: ≤1; missing data in 4.6%). The median observation period was 388.5 days (range 4–818).The median daily dose of NI was 600 mg (range 150–800 mg) with an initial NI daily dose of 600 mg in ~94% of the cases and a final NI daily dose of 600 mg >88% of the cases. There were 13.6% of pts with one and 3.2% with >1 interruption of therapy with a median of 14 days for the duration of each interruption (range 1–319). At last visit 80.2% (of 266 pts with a hematologic examination) had a CHR (10.7% with missing data), 81.5% (of 27 pts with a cytogenetic examination) had a CCyR (96.3% with PCyR), 52.3% (of 199 pts with a molecular examination) had an MMR (2% with missing data). In the subgroup of pts with molecular response (n = 178) the median time to MMR or better was 183 days (range 56–740). A premature study discontinuation took place in 16.1% of pts mostly due to AEs/non-hematologic toxicity, in 4 cases due to disease progression into blast crisis after development of a BCR-ABL mutation. Altogether, 76.7% of pts experienced AEs whereas 43.4% of those pts developed ≤3 AEs. Hematologic toxicity was observed in 9% of pts (6.5% with thrombocytopenia), non-hematologic toxicities occurred in 35.5% of pts. The most frequently reported AEs were skin reactions (pruritus 7.2%, rash 8.2%, alopecia 5.4%), fatigue (9%), headache (6.1%), gastrointestinal symptoms (nausea 6.8%, upper abdominal pain 6.1%, diarrhoea 5.4%), dizziness and dyspnoea (5%). No cardiac / vascular disorders were ≥ 2%. The most frequent biochemical abnormalities were increases in GGT (6.1%) and blood bilirubin (5%). Conclusions: These data from routine clinical management support the use of NI as an effective and safe drug for treatment of newly diagnosed Ph+ CML pts in CP. Disclosure: Bernd Lathan: No conflict of interest disclosed. Hans Tesch: Employment or Leadership Position: Niedergelassener Onkologe in der Hämatologisch-Onkologischen Gemeinschaftspraxis am AGAPLESION-Bethanien-Krankenhaus, Frankfurt; Advisory Role: Beratungstätigkeit u.a. Amgen, Astra Zeneca, Boehring Ingelheim, Celgene, Eisai, Genomic Health, GSK, Hexal, IMS-Health, Janssen-Cilag, Lilly, megapharm, Merck, Novartis, Onlopha, Pfizer, Roche, Sanofi-Aventis, Shire, Vifor; Financing of Scientific Research: Honorare u.a. Amgen, Astra Zeneca, Boehring Ingelheim, Celgene, Eisai, Genomic Health, GSK, Hexal, IMS-Health, Janssen-Cilag, Lapharm, Leo Pharma, Lilly, megapharm, Merck, Novartis, Onlopha, Pfizer, Roche, Sanofi-Aventis, Shire, Vifor. P457

Appearance disappearance of additional chromosome aberrations [+8, del(7q)] in Ph- cells in CML patient during treatment with TKIs of II generations, case-report Salvucci M.1,2,3, Zanchini R.1, Tonelli M.2, Giannini B.2, De Benedittis C.3, Valenti A.M.2, D’Addio A.1, Sensi A.2, Soverini S.3, Zuffa E.1, on behalf MPDsCML group of Romagna Local Health authority of Romagna, Ravenna, Italy, 2Medical Genetics Unit, Laboratory Services Centre, Cesena, Italy, 3Department of Experimental, Diagnosis and Specialty Medicine, Institute of Hematology ‘L. e A. Sèragnoli, University of Bologna, Bologna, Italy 1

Background: The occurrence of additional cytogenetic abnormalities (ACAs) in Philadelphia chromosome negative (Ph-) during tyrosine kinase inhibitors (TKIs) therapy for Chronic Myeloid Leukemia (CML) is a well-known event. Some abnormalities, including monosomy 7 and del(7q), have prognostic implications. We describe here the appearance of ACA clones, including a del(7q) in a patient with CP CML, who reached a late complete remission after frontline therapy with Nilotinib. Material and methods: A 46 year-old woman was admitted because of a painful splenomegaly on February 2012. According to clinic and hematologic conditions she was diagnosed a CP CML with a Sokal score of 1,75 (high risk). After an initial cytoreduction with Hydroxycarbamide, Nilotinib 300mg bid was started with poor tolerance due to hepatic and

Abstracts

hematologic toxicity. For this reason, the total dose of nilotinib, in the first 3 months never exceeded 75% of the programmed dose. Table I shows the cytogenetic and molecular data, according to ELN CML recommendations. Results and conclusions: A complete cytogenetic response (CCR) was achieved at month +18 and a major molecular response (MMR) at month +22, both in the peripheral blood and in the bone marrow. Point mutations were never detected all over the course of the disease. A trisomy 8 was observed in 20% of the Ph- metaphases at +12, in 60% at +14, and in 10% at +18. A del(7q) was observed in 5–10% of the Ph- metaphases between +14 and +25 months and no more thereafter. At +18, UDS analysis showed a 35 INS, this sequence variation consist in the retention of 35 nucleotides from intron 8 at the exon 8 to exon 9 border, it is a kinase inactive and should not play any role in TKI resistance. The patient, with a cumulative follow up of 37 months (19 months after the CCR and 15 after the MMR) is now in MMR (MR3) under nilotinib treatment. Ph- clones carrying trisomy 8 and del(7q) appeared and disappeared during the course of the disease, apparently with no adverse effect. As previously described, these abnormalities may be only the expression of genetic instability (confirmed by the detection of a 35 INS). Moreover, a long lasting CCR and MMR may be achieved also very late under nilotinib treatment. A strict molecular follow up is pursued. Disclosure: No conflict of interest disclosed. P458

Treating patients with Chronic Myeloid Leukemia (CML) in Chronic Phase (CP) with Dasatinib: PCR-monitoring, adherence, quality of life, therapy satisfaction (DasPAQT) Tesch H.1, Lange T.2, Hasford J.3, Belleville E.4, Anhuf J.5, Fietz T.6, Wolff T.7, Janssen J.8, Geer T.9, Steinmetz T.10 Hämatologisch-Onkologische Gemeinschaftspraxis am BethanienKrankenhaus, Frankfurt am Main, Germany, 2Asklepios Klinik Weißenfels, Klinik für Hämatologie und internistische Onkologie, Weißenfels, Germany, 3Institut für medizinische Informationsverarbeitung, Biometrie und Epidemiologie (IBE) der LMU, München, Germany, 4ClinSol GmbH & Co. KG, Würzburg, Germany, 5Onkologie Duisburg Nord, Duisburg, Germany, 6Schwerpunktpraxis für Hämatologie und Internistische Onkologie, Gastroenterologie, Singen, Germany, 7Onkologie Lerchenfeld, Hamburg, Germany, 8Gemeinschaftspraxis für Hämatologie und Onkologie, Westerstede, Germany, 9Diakonie-Klinikum Schwäbisch Hall GmbH, Klinik für Innere Medizin III, Schwäbisch Hall, Germany, 10 Praxis für Hämatologie und Onkologie, Köln, Germany 1

Introduction: Despite tyrosine kinase inhibitors (TKI) unprecedented efficacy in clinical trials, there is a lack of published data on how CML is managed in real-life clinical practice settings. This non-interventional study (NIS) is designed to collect real-life data on CML-treatment with Dasatinib in clinical routine. Emphasis lies on health care provided in office based physicians as most CML patients are treated outside of clinical studies. Methods: Main inclusion criteria are patients with newly diagnosed Ph+ CP-CML and CML patients in chronic phase resistant or intolerant to prior therapies, including Imatinib. Patients are treated with Dasatinib according to the clinical routine. Follow up is documented for a maximum of 24 months. A total of 300 patients from up to 75 centers (hematological and oncological hospitals or practices) will be recruited in order to answer the following questions: What is the pattern of use of Dasatinib in CML patients in real-life, including Dasatinib treatment strategies in first-line chronic CML or in a switch setting? What is the effectiveness and outcome of the treatment and what prognostic clinical and scientific factors determine the choice of treatment strategy? What is the patient-reported benefit and the impact of first-line Dasatinib CML treatment on patient quality of life? What are the rates of adherence/compliance, how satisfied are patients with their treatment, what patient-related factors lead to treatment discontinuation? Results: Baseline characteristics, treatment setting and treatment duration of the first 100 patients will be presented.

Oncol Res Treat 2015;38(suppl 5):1–270

135


CONTENTS AUTHOR INDEX

Conclusion: This NIS is intended to provide insight into the routine health care management of CML-patients treated with Dasatinib and its related outcomes. The factors that CML patients and treating physicians may encounter in a real-life setting will be observed to understand the benefits an effectiveness of Dasatinib CML treatment. Disclosure: Hans Tesch: Employment or Leadership Position: Niedergelassener Onkologe im Centrum für Hämatologie und Onkologie Bethanien Frankfurt.; Advisory Role: Beratungstätigkeit u.a. Novartis, Roche, Bristol-Myers Squibb; Financing of Scientific Research: u.a. Novartis, Roche, Bristol-Myers Squibb; Expert Testimony: Ja, Bristol-Myers Squibb. Tilmann Steinmetz: Employment or Leadership Position: Geschäftsführer X-Med GmbH, Leitung: Onkologie-Köln; Advisory Role: Amgen; BMS, Celgene, Hexal-Sandoz, Medice, Novartis; Janssen-Cilag; Pharmacosmos; Roche; Vifor; Stock Ownership: Kommanditist von GermanOncology; Financing of Scientific Research: KV-No, Privatpatienten; Expert Testimony: Studienleitung IIT/NIS/Register: Amgen, Celgene, Novartis, Vifor Studienarzt für Studien folgender Sponsoren: AIO, Amgen, BMS, Celgene, IoMedico, Janssen-Cilag, Kompetenznetz Maligne Lymphome und KN Akute Leukämien, Lilly, Merck, Novartis, Pfizer, Pharmacosmos, Ribosepharm, Roche, Schering, SKB, Wyeth,; Other Financial Relationships: Reisekosten: Alexion, Amgen, Bayer, Celgene, Janssen-Cilag, Novartis, Sanofi, Vifor; Immaterial Conflict of Interests: Mitgliedschaften: DGHO, ESMO, DGPM, BNHO, NIONo (Vorsitz)

Posterdiskussion Lungenkarzinom P459

Second line chemotherapy in small cell lung cancer – a retrospective analysis of two cancer centers Rieke J.1, Späth-Schwalbe E.2, Scholz C.W.3, Dieing A.3 Charité – Universitätsmedizin Berlin, Hämatologie und Onkologie, Berlin, Germany, 2Vivantes Klinikum Spandau, Klinik für Hämatologie und Onkologie, Berlin, Germany, 3Vivantes Klinikum Am Urban, Klinik für Hämatologie und Onkologie, Berlin, Germany 1

Background: Small Cell Lung Cancer (SCLC) has a high remission rate on first line treatment. However, most patients (pts) unfortunately relapse within a few months. Presenting frequently in a compromised performance status (PS) already at diagnosis, only a small part of these patients are eligible for second line therapy. To have a closer look at these patients as well as the clinical course in daily routine, we performed a retrospective analysis on SCLC pts treated 2nd line at two cancer centers in Berlin, Germany between 2008 and 2014. Methods: Of a retrospectively compiled database with 168 SCLC-patients receiving cytostatic therapy, we analyzed the patients treated with 2nd line chemotherapy regarding PS, comorbidity, type of therapy and survival. Comorbidity was assessed by the cumulative illness rating scale (CIRS). Results: Of 168 pts, 52 (31%) received 2nd line chemotherapy. Of those, 41 (79%) were male, compared to 68% of all SCLC patients. Median Karnofsky Performance Score (KPS) at diagnosis was 70% for all patients. In the group that received 2nd line therapy, KPS was 70% as well at diagnosis and at start of 2nd line therapy. Comorbidity score at diagnosis was 8 for all patients, and 9 for patients who received 2nd line therapy later on. Regimens used were ACO II or similar (cyclophosphamide, doxorubicin and / or vincristine)(n = 37; 71%) and topotecan (n = 15; 29%). Monotherapy was applied in 22 pts. (42%), whereas 30 pts (58%) received combination therapy. Overall Response Rate (ORR) was 19% e.g. 0 pts with CR and 10 pts with PR. In addition, in five patients (10%) SD was achieved, resulting in a disease control rate of 29% (15 pts). Median Survival from first diagnosis was 11.7 months, from start of 2nd line therapy 3.0 months. Conclusions: In our cohort, only patients who maintained a reasonable clinical performance received a 2nd line therapy. Overall response rate and OS from the beginning of the 2nd line treatment were only moderate in our cohort and shorter than data from published clinical trials, probably reflecting the positive selection in clinical trials.

136

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. P460

Incidence of driver mutations in lung adenocarcinomas: Rural versus industrialized environment Falk M.1, Baumhardt M.1, Griesinger F.2, Lüers A.2, Wiest G.3, Tiemann M.4 Institut für Hämatopathologie Hamburg, Molekularpathologie, Hamburg, Germany, 2Pius-Hospital Oldenburg, Hämatologie und Onkologie, Oldenburg, Germany, 3Asklepios Klinik für Pneumologie Harburg, Hamburg, Germany, 4 Institut für Hämatopathologie, Hamburg, Germany 1

Introduction: The incidence of driver mutations in adenocarcinomas of the lung depends on factors like gender, ethnicity, smoking status and exposure to mutagens. Here, we describe significant discrepancies of driver mutation frequencies in two different patient cohorts from northern Germany, one cohort originating from a rural area, the second from an urban, significantly industrialized environment. Methods: Patients with lung adenocarcinomas were genetically analyzed as part of our routine diagnostics from two major northern German lung centers, Pius Hospital Oldenburg and Asklepios Klinik Harburg. EGFR mutational analysis (Ex. 18–21) was performed by Cobas or direct Sanger sequencing. KRAS mutations (Ex. 2,3) were analyzed by Sanger Sequencing, ALK-translocations were tested by IHC, FISH or both. BRAF mutational analysis was performed by a "homebrew" allele specific realtime PCR and TP53 mutations by either Sanger- or Next Generation sequencing (NGS,454, Roche) . For statistical analysis fisher´s exact test was applied. Results: Altogether, 748 lung adenocarcinoma specimen from 2 different northern German lung centers were molecularly analyzed and data from both institutions were compared. The mean age in both cohorts was similar (66 vs. 68 years), while significantly more women were represented in the Oldenburg group (rural area). Although both patient cohorts originated from northern Germany, in the Oldenburg group TKI sensitive EGFR mutations were significantly overrepresented (14% vs. 6%, p < 0,0001). Along those lines, numbers of TKI sensitive ALK translocations were also significantly more frequent (5% vs. 1%, p = 0,0036). In contrast, TKI refractory KRAS mutations were slightly more prevalent in the AK Harburg cohort (industrialized area). TP53 mutations were present in 44% (Oldenburg) vs. 46% (Harburg). Further, we detected a significantly higher proportion of TP53 Exon 5 mutations in the Hamburg cohort, potentially indicative of increased tabacco abuse and/or exposure to industrial mutagenic substances (i.e. asbestos). Conclusion: We observed significant differences with regard to the frequency of targetable driver mutations in patients from two distict lung clinics in northern Germany. Since the geographical region is comparable, we speculate that environmental factors like work and living conditions (i.e. exposure to industrial mutagens) as well as socioeconomic status (i.e. smoking habits) might be part of the underlying reason. Disclosure: No conflict of interest disclosed. P461

RacGAP1 – a novel marker for tumour progression and invasion in bronchopulmonary neuroendocrine neoplasms? Specht E.1, Wirtz R.M.2, Kaemmerer D.3, Reimann C.1, Schmidt L.1, Nabian B.1, Sänger J.4, Lupp A.1 Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany, 2STRATIFYER Molecular Pathology GmbH, Köln, Germany, 3Department of General and Visceral Surgery, Zentralklinik, Bad Berka, Germany, 4Laboratory of Pathology and Cytology, Bad Berka, Germany 1

Introduction: Rac GTPase activating protein 1 (RacGAP1) belongs to the family of GTPase activating proteins, which interact with the active GTPbound form of small G proteins of the Rho family. They catalyse GTP hydrolysis, which leads to the transition into the inactive GDP-bound form of Rho GTPases. Here, RacGAP1 negatively regulates the actions of the Rho GTPases Rac1 and Cdc42, although it is an indirect activator of RhoA

Abstracts


CONTENTS AUTHOR INDEX

as well. Because it is a part of the centralspindlin complex, RacGAP1 is an important key player during the cytokinesis. Furthermore, there are references for its involvement in the adhesion, migration, differentiation and invasion of cells, whereby it became of interest in oncology. So far there are only few studies on RacGAP1, but all show significant increased expression levels in tumours of different localization in comparison to the normal tissue. For this reason, we wanted to assess the RacGAP1 expression in a series of bronchopulmonary neuroendocrine neoplasms (BPNEN) and different tumour cell lines to further characterize this protein. Methods: Formalin-fixed, paraffin-embedded specimen from 116 patients with BP-NEN, comprising 26 typical carcinoids, 30 atypical carcinoids, 52 small cell lung cancer (SCLC) patients (27 with good, 18 with bad prognosis; cut-off: survival </> = 30 months) and 8 patients with large cell neuroendocrine lung carcinomas, were quantified for their respective RacGAP1 protein and mRNA levels by means of immunohistochemistry and qRT-PCR. Expression levels were correlated with clinical data and other markers, like Ki-67, Chromogranin A, somatostatin receptors or the chemokine receptor CXCR4. Different tumour cell lines were characterized for RacGAP1 protein levels by Western blot analysis and immunocytochemistry. Results: RacGAP1 shows an increased expression to poorer differentiated and more aggressive tumour entities. It correlates with other proliferation markers and the CXCR4 levels. Tumours with increased RacGAP1 intensities in the invasive front have a poor prognosis. Similarly, SCLC with bad prognosis showed significant higher RacGAP1 protein and mRNA levels compared to those with good prognosis. Here, Ki-67 revealed no difference. Conclusions: RacGAP1 is a poorly characterized, but promising target in cancer diagnostics and maybe therapies with very strong prognostic implications. More studies should be conducted to better understand its role in carcinogenesis and cancer progress. Disclosure: No conflict of interest disclosed. P462

Molecular testing in NSCLC stage IV: Why are not 100% of patients tested? Lüers A.C.1,2, Neemann N.1, Hoheisel M.2, Wedeken K.2, Prenzel R.3, Scriba D.C.4, Willborn K.C.5, Stropiep U.2, Tiemann M.6, Griesinger F.1,2 Pius-Hospital, University Department Internal Medicine-Oncology, Oldenburg, Germany, 2Pius-Hospital, Hematology and Oncology, Oldenburg, Germany, 3 Pius-Hospital, Pneumology, Oldenburg, Germany, 4Pius-Hospital, Thoracic Surgery, Oldenburg, Germany, 5Pius-Hospital, Radiotherapy, Oldenburg, Germany, 6Hematopathology, Hamburg, Germany 1

Introduction: The current guideline for NSCLC recommends testing for molecular alterations in all non-squamous cell carcinoma patients stage IV regardless of smoking status and in squamous cell carcinoma patients if they are never or light smokers (<10 packyears, >15 years smoking cessation), with the aim to offer access to molecular stratified therapy. However, specifically in lung cancer, most patients are diagnosed in an in-patient setting for which testing is not yet reimbursed, which might create hurdles for the testing of all patients. Therefore, the intention of this study was to analyze the number of patients and the reasons for not testing in our lung cancer center which has access to the out-patient setting. Methods: All primary NSCLC cases stage IV in the lung cancer center were analyzed based on the patients’ files between 01/2014 and 03/2015 for the presence of testing results of molecular alterations and reasons for non-testing. Results: Between 01/2014 and 03/2015 159 primary cases of NSCLC stage IV were diagnosed at our lung cancer center. 132 patients received testing for molecular alterations EGFR, ALK, ROS (83%), 27 patients did not (17%). Out of these 27 patients, 10 patients (37%) continued treatment in an oncologic practice, and recommendation for testing was not followed. 6 patients (22%) died shortly after first diagnosis before molecular testing was initiated and before patients received therapy. 4 patients (15%) decided in favor of best supportive care. One patient with adenocarcino-

Abstracts

ma and a smoking history of 150 packyears (4%) was initially treated at a different hospital and has not progressed since starting chemotherapy, so molecular testing has not been performed as of yet. One patient (4%) was treated with chemotherapy with curative intention because of a single brain metastasis. Two patients (7%) were started on chemotherapy due to massive tumor load and symptoms, both patients died shortly after start of chemotherapy. Three patients (11%) were not tested because of no specific reasons. Conclusions: Even in an optimal setting of a certified lung cancer center only 83% of NSCLC stage IV that qualify for molecular testing receive it. Reasons why molecular testing is not performed are multifold. Special attention must be paid to these pitfalls in order to offer all patients the molecular stratified therapy. Reimbursement of testing in the in-patient setting will most likely improve test rates considerably. Disclosure: Anne Lüers: Advisory Role: Boeringer Ingelheim; Expert Testimony: Astra Zeneca. Frank Griesinger: Advisory Role: Boeringer Ingelheim, Roche, Pfizer, Astra ­Zeneca, Novartis, Lilly, MSD, BMS, Clovis; Expert Testimony: Astra Zeneca. P463

German country-wide surveys from 2012 and 2014 about EGFR mutational testing algorithms adopted by medical doctors in patients with NSCLC Ostermann H.1, Ukena D.2, Radke S.3, Hörnig S.3, Freitag A.4 Klinikum der Universität München – Großhadern, Medizinische Klinik III – Abteilung Hämatologie/Onkologie, München, Germany, 2Klinikum Bremen Ost, Klinik für Pneumologie und Beatmungsmedizin, Bremen, Germany, 3 AstraZeneca GmbH, Medical Affairs, Wedel, Germany, 4AstraZeneca GmbH, Wedel, Germany 1

Tyrosine kinase inhibitors (TKIs) targeting the epidermal growth factor receptor (EGFR) can significantly improve PFS for patients with advanced NSCLC harboring activating EGFR mutations (EGFRM+ aNSCLC) compared to chemotherapy. The German lung cancer guidelines recommend EGFRM testing for aNSCLC stage IIIB/IV before therapy. If an activating mutation is detected, an EGFR TKI is recommended as 1st line therapy. Two and four years after the 1st EGFR TKI (Gefitinib, 2009 in Europe) was approved for EGFRM+ aNSCLC, we studied whether physicians have implemented EGFRM testing into their clinical routine. We surveyed EGFRM testing in specialized lung clinics, academic and non-academic clinics and independent medical oncology offices. Two surveys recorded 1) frequency and selection criteria for EGFRM testing, 2) logistics of test initiation, histological and cytological sample preparation, and 3) barriers of EGFRM testing. In 2012, 60% of patients with aNSCLC stage IIIB/IV underwent an EGFRM analysis prior to treatment initiation. This number increased to 78% by the end of 2014. 12.5% and 11.7% of the patients were EGFRM positive in 2012 and 2014, respectively. Interestingly, in 2012, the lowest test rate of 38% was found in lung clinics but increased to 79% between surveys. Test rates had hardly changed in other institutions. Reasons against testing included cost, insufficient sample quantity and quality and pressure to treat. Selection criteria for testing were histology (~96%), smoking habits (~48%), gender (~27%), performance status (~22%) and age (~18%). A total of 11.3% and 8.9% of all patients were treated with EGFR TKIs as 1st line therapy in 2012 and 2014, respectively. In 2014, we added a sub-survey asking for the testing of KRAS, ALK and ROS1. ALK testing was most frequent with 53% followed by KRAS, 39%, and ROS1, 14%. Our surveys show that mutational analysis has already played an important role in 2012 for the treatment of NSCLC and that awareness for testing has increased recently. However, the data also indicate that there is still room for improvement regarding testing rate and the use of EGFR TKIs in line with German Lung Cancer Guidelines for EGFRM+ aNSCLC. Disclosure: Helmut Ostermann: Advisory Role: Teilnahme am Advisory Board 2015; Financing of Scientific Research: Für Teilnahme am Advisory Board und Posterpräsentation beim ELCC. Alexander Freitag: Employment or Leadership Position: Vollzeit-Angestellter.

Oncol Res Treat 2015;38(suppl 5):1–270

137


CONTENTS AUTHOR INDEX

P464

Influence of maintenance therapy on incidence of 2nd line therapy and OS in NSCLC IV Lüers A.C.1,2, Neemann N.1, Prenzel R.3, Scriba D.R.4, Hoheisel M.2, Wedeken K.2, Willborn K.C.5, Griesinger F.1,2 Pius-Hospital, University Department Internal Medicine-Oncology, Oldenburg, Germany, 2Pius-Hospital, Hematology and Oncology, Oldenburg, Germany, 3 Pius-Hospital, Pneumology, Oldeburg, Germany, 4Pius-Hospital, Thoracic Surgery, Oldenburg, Germany, 5Pius-Hospital, Radiotherapy, Oldenburg, Germany 1

Introduction: One of the strongest rationale for maintenance therapy in NSCLC is the fact that exposure to 2nd line therapy is only 40–60% in clinical trials in specialized treatment centers. Even with follow-up intervals of 6 weeks, the 2nd line treatment rate does not seem to increase. We analyzed the exposure of 2nd line therapy as well as OS and PFS in patients with stage IV NSCLC in the subgroups no 2nd line, 2nd line after maintenance and 2nd line without maintenance therapy. Methods: All primary lung cancer cases stage IV in the lung cancer center were analyzed based on the documentation files between 2009 and 2013. Patients were followed-up between 1st and 2nd line therapy every 6–8 weeks according to S3 guidelines. Patients with EGFR+, ALK+ or ROS1+ were excluded from the analysis. Results: 221 patients were diagnosed with NSCLC IV (UICC7), or had systemic relapse of localized disease and were treated with 1st line therapy for metastatic disease. Of these, 160 (72%) received 1st line combination therapy with Carboplatin, 50 (23%) with Cisplatin and 11 (5%) with platin-free single agent therapy. 45 (19%) of all patients received maintenance therapy, most of them with bevacizumab. Of 221 patients, 203 (92%) progressed after 1st line therapy or 1st line and maintenance therapy. 106/163 (65%) of non-maintenance therapy patients received 2nd line therapy, 57 patients (36%) did not. Of 40 patients receiving maintenance therapy and requiring 2nd line therapy, 31 (78%) received 2nd line therapy. Reasons for not obtaining 2nd line therapy were captured and were manifold. Survival analyses showed significant differences regarding overall survival (median survival 21 (maintenance and 2nd line) vs. 13 (1st and 2nd line) months) but no relevant differences regarding progression free survival on 2nd line (median 2 months). Conclusion: In a certified lung cancer center and stringent follow-up every 6 to 8 weeks, 1/3 of patients do not receive 2nd line therapy because of various reasons. The application of maintenance therapy raises the chances of receiving 2nd line therapy and increases overall survival whereas progression free survival is not affected. Disclosure: Anne Lüers: Advisory Role: Boehringer Ingelheim; Expert Testimony: Astra Zeneca. Frank Griesinger: Advisory Role: Boehringer Ingelheim, Roche, Pfizer, Astra Zeneca, Novartis, Lilly, MSD, BMS, Clovis; Expert Testimony: Astra Zeneca. P465

The phase II ASCEND-7 (CLDK378A2205) trial: Ceritinib in patients (pts) with ALK-rearranged (ALK+) Non-Small Cell Lung Cancer (NSCLC) metastatic to the brain and/or leptomeninges Wolf J.1, Schneider C.-P.2, Potzner M.3, Cazorla Arratia P.4, Shen J.4, Branle F.5, von Pawel J.6 Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany, 2Zentralklinik Bad Berka, Abteilung für Internistische Onkologie und Hämatologie, Bad Berka, Germany, 3Novartis Pharma GmbH, Nürnberg, Germany, 4Novartis Pharmaceuticals Corporation, East Hanover, United States, 5Novartis Pharma AG, Basel, Switzerland, 6Asklepios Klinik für Pneumologie, Abteilung Onkologie, München-Gauting, Germany 1

Introduction: In pts with ALK+ NSCLC receiving the ALK inhibitor (ALKi) crizotinib (CRZ), disease progression often occurs within 1 year with the brain/central nervous system (CNS) as a common site of progression and relapse. The oral ALKi ceritinib showed a 20-fold greater potency than CRZ in enzymatic assays and crosses the blood-brain barrier with

138

Oncol Res Treat 2015;38(suppl 5):1–270

good CNS penetration in preclinical studies. In the pivotal phase I study (NCT01283516) ceritinib was highly active in ALK+ NSCLC pts regardless of prior CRZ exposure and achieved intracranial responses in 7 of 14 pts with measurable baseline brain lesions. Adverse events profile in these pts was similar to that of the full study population. Methods: This prospective phase II study is designed to evaluate the antitumor activity of ceritinib in pts with ALK+ NSCLC metastatic to the brain or leptomeninges. Eligible pts must have investigator assessed ALK+ NSCLC metastatic to the brain and ≥1 extracranial measurable lesion (RECIST v1.1). Pts must be neurologically stable ≥1 week prior to ceritinib and will be allocated to 1 of 5 cohorts: ARMS 1–4 (pts w/ active* brain mets, w/o LC)

Prior ALKi

NO prior ALKi treatment

Prior brain radiotherapy (BRT)

ARM 1

ARM 3

NO prior BRT

ARM 2

ARM 4

ARM 5: pts with leptomeningeal carcinomatosis (LC) with or without evidence of active lesion at baseline. *Lesion free of local treatment (stereotactic or Whole BRT) or lesions in unequivocal progression after radiotherapy. Ceritinib will be dosed 750 mg/day on a continuous schedule; study assessments are consistent across cohorts. Primary and key secondary objectives are whole body overall response rate and disease control rate, respectively. Other secondary objectives include intracranial and extracranial responses for all pts and for each cohort 1–4; overall survival and safety for all pts and for each cohort 1–5; and ceritinib pharmacokinetics in all pts. Disclosure: Jürgen Wolf: Advisory Role: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Financing of Scientific Research: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Expert Testimony: Bayer, Boehringer-Ingelheim, Novartis, Pfizer, Roche. Joachim von Pawel: Advisory Role: AbbVie, BMS, Daiichi Sankyo, Novartis, Pfizer. P466

Intercalated TKI and chemotherapy induction in EGFR mt+ NSCLC stage IIIA and IIIB: Report of 3 cases with complete pathologic remission in mediastinal lymph nodes Griesinger F.1,2, Lüers A.C.1,2, Falk M.3, Conradi I.3, Reinhardt M.4, Kluge A.5, Willborn K.C.6, Prenzel R.7, Scriba D.8, Henke R.P.9, Eberhardt W.E.E.10, Hallas C.3, Tiemann M.3 Pius-Hospital, University Department Internal Medicine-Oncology, Oldenburg, Germany, 2Pius-Hospital, Hematology and Oncology, Oldenburg, Germany, 3 Hematopathology, Hamburg, Germany, 4Pius-Hospital, Nuclear Medicine, Oldenburg, Germany, 5Pius-Hospital, Diagnostic Radiology, Oldenburg, Germany, 6Pius-Hospital, Radiotherapy, Oldenburg, Germany, 7Pius-Hospital, Pneumology, Oldenburg, Germany, 8Pius-Hospital, Thoracic Surgery, Oldenburg, Germany, 9Pathologie, Oldenburg, Germany, 10University Hospital DuisburgEssen, Oncology, Essen, Germany 1

Background: EGFR TKI treatment is standard of care in patients with metastasized NSCLC carrying an activating EGFR mutation. However, induction concepts in locally advanced NSCLC with EGFR mutation including TKI have not been studied extensively. Recently new focus has been shed on intercalated regimens of chemotherapy and TKI, showing improved PFS and OS. This concept was used as induction regimen in 3 patients with activating EGFR mutation in stages IIIA and IIIB. Methods: Patients were diagnosed according to standard imaging, histology and immunohistology methods. EGFR, KRAS, BRAF, ALK and P53 mutation analysis were performed with standard procedures. Remission induction was measured by RECIST 1.1. Results: 2 female never smokers (pt #1 and 3), ,and 1 male light smoker (pt#2) (5 py) were diagnosed with NSCLC, 2 with exon 21 L858R (#2,3) and 1 with Exon 19 deletion (#1). All patients carried a p53 mutation. Tumor stage was T3 (extension to mediastinal pleura) N2(2R,4R)M0, IIIA4

Abstracts


CONTENTS AUTHOR INDEX

(#1), T2aN3(4L,7,2R)M0 IIIB (#2) and T2N3M0. . Induction therapy was started with erlotinib 150 mg/d days -12 to -1 (#1,2) and gefitinib (#3) in order to prove responsiveness of the tumour to EGFR-TKI. On day 0 partial response or no progression was achieved in all patients. Therapy was continued with 3 cycles of dDocetaxel 75 mg/m2 d1 and cisplatin 50 mg/m2 d 1 and 2 qd22 in combination with erlotinib d4–19 (#1), 1 cycle of ddocetaxel and ccisplatin followed by 2 cycles of paclitaxel and carboplatin (#2) and switch from erlotinib to gefitinib with cycle 2 (#2) because of diarrhea and 3 cycles of docetaxel and cisplatin with gefinitib 250 mg d4–19 (#3). PR was achieved after 2 cycles in all patients. All patients were resected and regression grade IIB was remarked in mediastinal lymph nodes (#1–3), regression IIA was remarked in the primary tumor in 2 patients (#2,3), regression grade III in 1 patient (#1). All patients received adjuvant radiotherapy. Patients #1 and 3 are in CR, patient 2 developed one isolated CNS metastasis which has been stereotactically irradiated. No additional therapy, including TKI was administered postoperatively. Conclusion: Intercalated TKI treatment is a promising treatment in patients with EGFR mt+ locally advanced NSCLC. A phase II trial (NeoIntercal) trial is currently under way in 9 German centers in stages II and III using gGefitinib in combination with induction taxane based chemotherapy, supported by ASTRA Zeneca. Disclosure: Frank Griesinger: Advisory Role: Boehringer Ingelheim, Clovis, Astra Zeneca, MSD, Merck, Novartis, Pfizer; Expert Testimony: Astra Zeneca. Markus Tiemann: No conflict of interest disclosed. P467

Fig. 1. Initial CT scan before treatment.

85-year old patient with EGFR mutated metastatic adenocarcinoma of the lung – Excellent clinical benefit from treatment with erlotinib Schmid T.1, Köberle D.1, Buess M.1 St. Claraspital, Onkologie, Basel, Switzerland

1

Introduction: As shown in several clinical trials targeted therapies are the preferred treatment option for molecularly selected metastatic carcinomas of the lung due to their efficacy and patients’ tolerance. However, patients over 80 years old are under-represented in these trials and in daily-practice do still not undergo complete diagnostic work-up at many places. Methods: We report on an 85-year old patient with relapsed metastatic adenocarcinoma of the lung, 8 months after right sided lobectomy. Her performance status declined within a few weeks to a score of 3. She was tachydyspnoeic when she was referred to discuss best supportive care. Before further counselling, molecular testing revealed an EGFR mutation in Exon 19 (deletion p.E746-A750). We initiated a therapy with erlotinib. Results: After two weeks of treatment, the patient’s quality of life already improved and a CT scan after 3 months showed a very good partial remission. Erlotinib was tolerated at a dose of 150mg/d without any relevant toxicity. 12 months later she is still being treated without any sign of tumor progression and with an excellent quality of life. Important to her, she is even able to take care of her spouse. Conclusions: This elderly patient with an initially poor performance status gained optimal palliative benefit from erlotinib. Molecular testing for EGFR mutations and EML4-ALK translocations should be performed in all patients with metastatic non-squamous non-small-cell lung cancer according to current guidelines. In the near future most likely additional predictive genetic alterations will be tested. Elderly and fragile patients who are not candidates for chemotherapy should no longer be excluded from complete diagnostic work-up, as they can benefit from targeted therapies with acceptable side-effects.

Abstracts

Fig. 2. CT scan after 3 months of treatment with erlotinib. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

139


CONTENTS AUTHOR INDEX

P468

Induction therapy with intercalated TKI and chemotherapy in NSCLC with activating EGFR mutation in stages II-IIIB: NeoIntercal Griesinger F.1, Sebastian M.2, Serke M.3, Büttner R.4, Waller C.5, Reinmuth N.6, Graeven U.7, Lüers A.1, Radke S.8, Karatas A.9, Heukamp L.10, Tiemann M.10, Overbeck T.11 Pius-Hospital Oldenburg, Hämatologie/Onkologie, Oldenburg, Germany, Universität Frankfurt am Main, Med. Klinik II, Hämatologie/Onkologie, Frankfurt, Germany, 3Lungenfachklinik Hemer, Pneumologie III, Hemer, Germany, 4Universitätsklinikum Köln AöR Institut für Pathologie, Köln, Germany, 5Universität Freiburg – Klinik für Innere Medizin I, Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 6Lungclinic Grosshansdorf, Grosshansdorf, Germany, 7Kliniken Maria Hilf GmbH – Innere Medizin I, Mönchengladbach, Germany, 8AstraZeneca GmbH, Medical Affairs, Wedel, Germany, 9AIO gGmbH, Berlin, Germany, 10Hematopathology Hamburg, Hamburg, Germany, 11Universitätsmedizin Göttingen, Hämatologie/Onkologie, Göttingen, Germany 1 2

EGFR TKI treatment is standard of care in patients with metastasized (m) NSCLC carrying activating EGFR mutations. 1st and 2nd generation agents lead to response rates of up to 70%. Recently, new focus has been shed on intercalated regimens of chemotherapy and TKI showing improved PFS as well as OS in an unselected Asian population (Wu et al. 2013). Response is a predictor of PFS and OS in limited and locally advanced NSCLC. Chemotherapy induction alone leads to pCR rates of no more than 15%. No data have been generated for induction therapy including EGFR TKI in EGFRM+ NSCLC. Four cases treated in one center have shown the feasibility and tolerability of an intercalated induction therapy concept (Lüers et al. 2013). NeoIntercal, a single arm phase II study has been initiated in 9 centers in Germany. Patients with stage II to IIIB staged according to local center standards will be screened for EGFR mutations by certified pathologists. EGFRM+ patients will receive gefitinib 250 mg / die p.o. on d-12 to -1 (d1 = first day of first cycle of chemotherapy) followed by 3 cycles of taxane and platin containing chemotherapy with intercalated gefitinib on d4-d20 of each cycle. After 2 cycles, restaging CT will be performed and surgery will be scheduled during the 4th or 5th week of the last cycle of CTx-gefitinib. Pathologic response rate is the primary endpoint. If more than 30% of patients achieve pCR (regression grades IIB and III according to Junker) in the mediastinal lymph nodes, the 2nd part of the study is planned to enroll an additional 28 patients. Secondary endpoints include OS, PFS, relapse rate, toxicity and feasibility. A liquid biopsy project is included in the study to correlate tumor biopsy results with liquid biopsy results and to monitor therapy effects. Study preparation and trial center recruitment are completed and the enrollment of the first patient is planned for 3Q2015. An interim analysis will be performed approximately 12 months after enrollment initiation with data from 21 patients. The study is scheduled to end by 2019 with a total of 49 patients (follow up period of 24 month) should the interim analysis be positive. According to our knowledge, NeoIntercal is the first study in the neoadjuvant setting with curative intent applying an intercalating combination of chemotherapy and targeted therapy. The NeoIntercal study group believes that this study will potentially contribute to the improvement of EGFRM+ NSCLC therapy.

Posterdiskussion

Lymphatische Neoplasien und nicht-maligne Hämatologie P469

Validation of the EBMT risk score in a cohort of German ALL patients Fürst D.1,2, Arnold R.3, Zollikofer C.1,2, Tsamadou C.1,2, Schrezenmeier H.1,2, Mytilineos J.1,2 Institut für Klinische Transfusionsmedizin und Immungenetik, Universität Ulm, Transplantationsimmunologie, Ulm, Germany, 2DRK-Blutspendedienst BadenWürttemberg-Hessen, Ulm, Germany, 3Charite Universitätsmedizin Berlin, Hämatologie / Onkologie, Berlin, Germany 1

Introduction: The EBMT risk score has been proposed for risk assessment of patients undergoing allogenic stem cell transplantation. The risk score has been established on the EBMT database including cases between 1980–2005. We validated the EBMT risk score in a cohort of acute lymphocytic leukemia (ALL) patients (n = 473) transplanted with unrelated stem cell transplantation at German centres between 1997 and 2012. Patients and methods: Only first allo-transplants were included. Most of the transplantations in the validation dataset (n = 309, 65.5%) have been performed between 2006 and 2012. For each patient the EBMT risk score was calculated. Risk scores from 1 to 6 were observed. Results: Most patients fell into the categories: Score 2 (n = 123, 26%), score 3 (n = 150, 31.7%) and score 4 (n = 96, 20.3%). Using this score a cox-regression model was fitted to obtain risk estimates for each score level. Risk estimates were for Score 2: HR 1.47 (CI 0.70–3.11, p = 0.308), for score 3: HR 1.82 (CI 0.87–3.79, p = 0.112), for score 4: HR 2.81 (CI 1.34–5.90, p = 0.006), for score 5: HR 4.01 (CI 1.88–8.57, p < 0.001) and for score 6: HR 5.82 (CI 2.46–13.74, p < 0.001). Conclusions: We validated the EBMT risk score in a cohort of German patients transplanted for ALL. Most of the transplantations in our dataset could not have been included in the original dataset used for description of the EBMT risk score as they were performed in more recent years. Our dataset confirms the validity of the EBMT risk score. Tab. 1. Regression estimates for different levels

HR

2.5% CI

97.5% CI

p-Value

Score 1

1.00

Score 2

1.47

0.70

3.11

0.308

Score 3

1.82

0.87

3.79

0.112

Score 4

2.81

1.34

5.90

0.006

Score 5

4.01

1.88

8.57

<0.001

Score 6

5.82

2.46

13.74

<0.001

Disclosure: Frank Griesinger: Advisory Role: Teilnahme an Advisory Boards; Financing of Scientific Research: Für wissenschaftliche Beratungen und Präsentationen. Tobias Overbeck: No conflict of interest disclosed.

140

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

P471

Quality of life (QoL) assessment in patients allografted for poor-risk chronic lymphocytic leukemia (CLL) Hahn M.1, Dietrich S.1, Hegenbart U.1, Bondong A.1, Ho A.D.1, Dreger P.1 Universität Heidelberg, Medizinische Klinik V, Heidelberg, Germany

1

Fig. 1. Survival functions of different levels o. Disclosure: No conflict of interest disclosed. P470

IRF8 mutation screening in different types of leukemia reveals an inactivating IRF-8 mutation in a young patient with high risk pro-B acute lymphatic leukemia Chifudov S.1, Burmeister T.2, Stilgenbauer S.3, Burchert A.4 Universitätsklinikum Giessen und Marburg GmbH, Klinik für Hämatologie, Onkologie und Immunologie, Marburg, Germany, 2Charité Berlin, Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany, 3Universitätsklinikum, Ulm, Germany, 4Universitätsklinikum Marburg, Hämatologie/Onkologie und Immunologie, Marburg, Germany 1

Introduction: The transcription factor interferon regulatory factor 8 (IRF8) governs hematopoietic stem cell differentiation into monocytes / dendritic cells as well as B-cells. In humans, IRF8 malfunction may be due to loss of expression or point mutation. In line with the phenotype of IRF8 knockout mice, which display a myeloproliferative syndrome and Th1-immundeficiency, IRF8 loss of function in humans has been linked to myelodysplasia, myeloid leukemogenesis, and rare immunological disorders. Methods: Here we investigated patients with acute lymphatic leukemia (ALL) (n = 59), chronic lymphatic leukemia (CLL) (n = 52) and several patients with mantle cell lymphoma (MCL) for mutations in IRF8 using Sanger sequencing Results: We identified two previously published missense mutations (F328V and Y242H) in two patients with ALL and MCL, respectively. Another young female patient with high risk pro-B-ALL was found to display a previously not reported heterozygous p.R296H mutation in the IRF association domain (IAD) of IRF8. The IAD is known to control IRF8 protein interaction and thus transcriptional target selection. Interestingly, the R296H mutation affects the same conserved amino acid of IRF8 as the one mutated in the BXH2 mice strain (Turcotte et al., JEM 2005). BXH2 mice develop a myeloproliferation and immunodeficiency at early age, but only in the case of a homozygous mutation. The mutation pR296H was associated with a higher IRF8 mRNA expression compared to pro-B-ALL samples with IRF8 wild type. Conclusion: IRF8 mutations may contribute B-cell leukemogenesis. Disclosure: No conflict of interest disclosed.

Abstracts

Introduction: Little is known about quality of life (QoL) of survivors of allogeneic hematopoietic stem cell transplantation (HSCT) for CLL. Patients and methods: In a single center cross-sectional analysis, QoL and employment status were assessed in patients (pts) allografted for poor-risk CLL. Eligible were all living pts who had undergone HSCT for CLL at our institution between 05/2005 and 04/2013. Eligible pts (n = 53) were asked to complete the FACT-BMT questionnaire, a validated measure of QoL in HSCT patients, considering 5 dimensions of QoL: physical (PWB), social (SWB), emotional (EWB) and functional well-being (FWB), and HSCT-specific aspects (bone marrow transplantation subscale, BMTS). An extra item was added to survey the employment status. Results: 40 questionnaires returned. Of the 40 pts at a median age of 59y (43–72), 28 (70%) were male, 32 (80%) had an unrelated donor (UD), 9 (23%) a mismatch donor. At QoL assessment, 34 (85%) were in CR and 14 (35%) had cGvHD. The median time from HSCT to QoL assessment was 50 months (9–98). Of 31 pts that reported about employment, 11 (28%) had retired or were unemployable >1 year before HSCT. Of the remaining 20 pts, 15 (75%) were fulltime, and 2 (13%) part time employed after HSCT. The median time from HSCT to fulltime employment was 10 mths (2–33). The median subscale scores were PWB 25/28, SWB 24/28, EWB 20/24, FWB 21/28, BMTS 31/40, resulting in a median FACT-BMT total score of 122/148 (range 87–144/148). This implies that half of the pts reported a good to excellent, the other half an intermediate to good aggregated QoL at assessment. The total score tended to be lower in pts unemployed after HSCT, MRD+ 12 months after HSCT, not in CR at QoL assessment, or that had progressive disease, transplanted from an UD or mismatch donor, with cGvHD history or cGvHD at QoL assessment, and in pts still on systemic immunosuppression (IS) 12 mo after HSCT. However, none of the scoring differences was significant with the patient number studied here. In pts that were fulltime employed after HSCT, FWB subscale score was significantly higher (p = 0.0265). Age, time to IS tapering and the number of previous therapies did not significantly affect the scoring. Conclusion: This study provides preliminary evidence that QoL after HSCT for CLL is perceived as acceptable or favorable in the vast majority of patients. A large proportion resumed fulltime employment after HSCT, which significantly affected functional well-being. Disclosure: No conflict of interest disclosed. P472

Favorable immune signature in CLL patients, defined by antigen-specific T-cell responses, might prevent secondary skin cancers Stickel J.S.1, Kowalewski D.J.2, Schuster H.2, Kahn S.1, Backert L.2,3, Kanz L.1, Salih H.R.1,4, Rammensee H.-G.2,5, Stevanovic S.2,5 University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany, 2Interfaculty Institute of Cell Biology, Department of Immunology, Tübingen, Germany, 3University of Tübingen, Department of Computer Science, Tübingen, Germany, 4German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Clinical Collaboration Unit Translational Immunology, Heidelberg, Germany, 5DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Tübingen, Germany 1

The course of chronic lymphocytic leukemia (CLL) predisposes patients to the development of secondary malignancies, which might be due to a generalized immunosuppression induced by the disease and/or the therapies used to treat the disease. Skin cancer, including melanoma, is the most common secondary malignancy reported in these patients.

Oncol Res Treat 2015;38(suppl 5):1–270

141


CONTENTS AUTHOR INDEX

In a recent study (Kowalewski et. al., PNAS 2015) we reported on spontaneous memory T-cell responses against CLL-associated antigens defined by HLA ligandome analysis in CLL patients. Moreover, retrospective analysis suggests a significant survival benefit for CLL patients showing immune reactions to more than one of the tested antigens. Here we analyzed this CLL patient cohort for the incidence of secondary malignancies, and in particular for skin cancers. 45 CLL patients (Binet A n = 15; B, n = 15; C n = 15, 33.3% (15/45) with prior CLL specific therapy) were analyzed for antigen-specific T-cell responses in IFNγ-ELISPOT assays. 13 patients (28.9%, response cohort) showed > 1 antigen specific T-cell responses, while 32 patients (71.1%, negative cohort) showed none or only one immune response. 40.0% (6/15) of CLL patients with Binet stage A and 33.3% (5/15) with stage B were classified as responders, whereas only 13.3% (2/15) of patients with stage C showed >1 immune responses. Of the included CLL patients with prior therapy only one showed > 1 immune responses. In the analyzed CLL patient cohort 7 skin cancers (melanoma n = 2, squamous cell carcinoma n = 2, basal cell carcinoma n = 3), 3 precancerous skin lesions and 9 other secondary malignancies including renal cell carcinoma (n = 2), colon (n = 3), breast (n = 2) and prostate cancer (n = 2) were observed. In the negative cohort 34,4% (11/32) of the patients developed a secondary malignancy, while only 15,4% (2/13) in the immune response cohort presented with secondary neoplasms. Strikingly all skin cancers and precancerous skin lesions were observed in the non-immune responder cohort. Furthermore multiple secondary malignancies were only observed in the cohort that showed no immune response. Taken together these data suggest that a favorable immune signature in CLL patients that is able to generate T-cell responses against CLL-associated antigens not only is associated with improved overall survival but also might protect these patients from the development of secondary skin cancers. Disclosure: No conflict of interest disclosed.

of the safety profile of RM therapy and an analysis of treatment modalities during induction and RM treatment. Results: From Oct 2009 to Mar 2012, 138 centers recruited 505 pts. 483 pts were evaluable for the final analysis. 310 pts received 1L induction therapy, and 173 pts were treated for relapse. Induction therapy led to CR in 47.7% and 42.8% of pts who received R as 1L or REL treatment, respectively. The 2Y PFS rate for the 1L pts (n = 305) was 88.3%; 95%CI: 84.0–92.6 (2Y OS rate: 96.9%; 95% CI: 94.7–99.1), and for the REL pts (n = 172) 76.0%; 95%CI: 68.8–83.3 (2Y OS rate: 95.4%; 95% CI: 91.8–99.1). At the end of RM the 1L group presented 58.4% of pts with CR, 21.0% with PR, 12.1% with stable disease (SD) and 8.5% with progressive disease (PD) at the end of RM. In the REL group a lower percentage of 51.2% achieved CR, whereas the percentage of PD (18.6%) was higher. The proportion of PR (20.9%) and SD (9.3%) was similar to the findings in 1L pts. Based on a preliminary safety analysis, adverse (serious) events were observed in 30.4% (14.1%) of the pts (drug-related in 13.5% (5.3%)) (n = 490). Most common drug-related adverse events (incidence ≥5%) of any grade were leukopenia (20.4%), neutropenia, bacterial infection (9.0%, both) and dyspnoe (5.4%). Conclusions: The results of our study confirm the previously reported efficacy and safety data of RM treatment in clinical practice. Disclosure: Ulrich Dührsen: Financing of Scientific Research: Roche Pharma AG; Expert Testimony: Roche Pharma AG. Christian Lerchenmüller: No conflict of interest disclosed. P474

A gain-of-function mutation in the Plcg2 gene protects mice from Helicobacter felis-induced gastric MALT lymphoma Huynh M.Q.1,2, Goßmann J.1, Stolte M.3, Lohoff M.4, Yu P.5, Finkernagel F.6, Garn H.7, Bittner A.1, Neubauer A.1 Uniklinik Marburg, Hämatologie, Onkologie und Immunologie, Marburg, Germany, 2Landeskrankenhaus Bregenz, Innere Medizin, Bregenz, Austria, 3 Krankenhaus Kulmbach, Pathologie, Kulmbach, Germany, 4Universität Marburg, Institut für Mikrobiologie und Hygiene, Marburg, Germany, 5Universität Marburg, Institut für Immunologie, Marburg, Germany, 6Universität Marburg, Institut für Molekularbiologie und Tumorforschung, Marburg, Germany, 7 Uniklinik Marburg, Institut für Labormedizin und Pathobiochemie, Marburg, Germany 1

P473

Application of Rituximab in Maintenance (RIM) therapy in Follicular Lymphoma (FL) in routine clinical practice – final results of a non-interventional study (NIS) in Germany Dührsen U.1, Dieing A.2, Reiser M.3, Prange-Krex G.4, Jentsch-Ullrich K.5, Lück A.6, Porowski P.7, Schwinger U.8, Broszeit-Luft S.9, Klawitter S.10, Krumm K.10, Lerchenmüller C.11 Universitätsklinikum Essen, Klinik für Hämatologie, Essen, Germany, Vivantes MVZ Am Urban, Berlin, Germany, 3Praxis internistischer Onkologie und Haematologie, Köln, Germany, 4Onkologie Praxis, Dresden, Germany, 5 Schwerpunktpraxis für Hämatologie und Onkologie, Magdeburg, Germany, 6 Zentrum für Onkologie und Urologie, Rostock, Germany, 7Onkologische Schwerpunktpraxis, Heilbronn, Germany, 8Internistische Schwerpunktpraxis Onkologie, Hämatologie, Gastroenterologie, Stuttgart, Germany, 9Onkologie Praxis, Lehrte, Germany, 10Roche Pharma AG, Grenzach-Wyhlen, Germany, 11 Gemeinschaftspraxis für Hämatologie und Onkologie (GEHO), Münster, Germany 1 2

Introduction: The standard of care for patients (pts) with advanced stages of FL requiring therapy is Rituximab (R) plus chemotherapy (CT) followed by R maintenance (RM) which demonstrated significant clinical benefit in both relapsed (REL) pts and after first-line (1L) induction. Standard treatment includes 6–8 cycles of R plus CT followed by a single infusion of RM every 2 or 3 months for 2 years (2Y). To assess the efficacy, safety and treatment regimens of RM in clinical practice this prospective, multicenter RIM NIS (referred to as Study ML22283) conducted in Germany was performed. Methods: The study included adult pts with previously untreated, relapsed or refractory FL, who were scheduled for RM therapy after achieving complete or partial response (CR or PR) to R-containing induction therapy. Treatment decisions were made by the treating physician prior to inclusion. Key parameters of efficacy were rate of progression-free (primary) and overall survival (PFS and OS) after 2Y of RM therapy, calculated by using the Kaplan Meier method. Further objectives were the evaluation

142

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: H. pylori-induced gastric MALT lymphoma development is dependent on different virulence factors of H. pylori strains and in part from the host´s genetic background. Phospholipase C gamma 2 (PLCG2) is important for B-cell survival and proliferation. We used BALB/c mice with a gain-of-function mutation in the Plcg2 gene (Ali5) to analyze its role in the development of gastric MALT lymphoma. Methods: BALB/c Plcg2Ali5/+ and wildtype (WT) mice were infected with H. felis and observed up to 16 months for development of MALT lymphomas. To assess proinflammatory cytokines in peripheral blood we used a RT PCR Array. Blood sera were collected to determine H. felis-specific immunoglobulins by ELISA. Mouse spleen mononuclear cells (MNCs) were used to assess CD73 expressing Tregs by flow cytometry. Results: We have found that H. felis-infected Plcg2Ali5/+ mice showed significantly less gastric MALT lymphomas compared to their WT littermates after 16 months of infection. These results were somehow unexpected, because we had assumed that Plcg2Ali5/+ mice might show a stronger gastric inflammatory response and thus an increased induction of MALT lymphomas after H. felis infection. This seemed likely, as Plcg2 activates NF-κB. Reduced MALT lymphoma development in H. felis-infected Plcg2Ali5/+ mice was due to downregulation of several proinflammatory cytokines, including two important inflammatory cytokines, IFNγ and IL1a. In addition, infected Plcg2Ali5/+ mice showed less H. felis-specific IgG1 (P = .0307) and IgG2a (P = .0016) antibody responses. It was previously shown that CD73 expressing Tregs suppress H. felis-induced gastritis in mice. We could show that Plcg2Ali5/+ mice possessed a significantly higher percentage of CD73+ Tregs compared to their WT littermates (P = .0017).

Abstracts


CONTENTS AUTHOR INDEX

Conclusions: These results show a blunted immune response of Plcg2Ali5/+ mice towards H. felis infection. Intriguingly, Plcg2Ali5/+ mice possessed higher numbers of CD73 expressing Tregs that might suppress the immune response to Helicobacter infection. We suggest that Plcg2Ali5/+ mice may be protected from developing gastric MALT lymphomas as a result of elevated Treg numbers, reduced response to H. felis and decrease of proinflammatory cytokines. Disclosure: No conflict of interest disclosed. P475

Waldenström’s macroglobulinemia as presented with specific cutaneous manifestations Műzes G.1, Csomor J.2, Sipos F.1 Semmelweis University, 2nd Department of Internal Medicine, Budapest, Hungary, 2Semmelweis University, 1st Department of Pathology and Experimental Oncology, Budapest, Hungary 1

Introduction: Waldenström’s macroglobulinemia (WM) represents a malignancy of lymphoplasmacytic (LP) cells, belonging to group of indolent B-cell NHLs. WM is a rare form of NHLs (incidence: ~3.1/1.000.000/yr), characterized mainly by intertrabecular infiltration of the bone marrow by clonal plasmacytoid cells with a specific immunophenotype, and by monoclonal IgM gammopathy. Though several, non-specific skin lesions could related to WM, the presence of specific cutaneous manifestations (i.e. skin infiltration by abnormal B-cells, and/or skin deposits of the paraprotein) is infrequent (1–3%). Materials and methods: Here we present an unusual case of WM with both specific cutaneous lesions. Results: The 68-year-old female patient with a past history of hypertension was presented to our Department in 2005 for evaluation of dark-bluish confluent plaques over her cheeks and bridge, affecting also the ears and shoulders with a duration of 2–3 yrs. (Earlier she was diagnosed by dermatologists with rosacea, later with contact dermatitis, and then with malar rash.) Routine laboratory tests indicated increased ESR and a mild anemia. Despite the ANA positivity her skin lesions were atypical for SLE. Upon skin biopsy from the left shoulder histological examination revealed deep, diffuse lymphoid infiltration of the dermis and the dermo-epidermal junction, suggestive for NHL/LPL. Further investigations, including serum immunofixation electrophoresis, and histopathologic and immunohistochemical analyses of crista biopsy specimen unequivocally proved the diagnosis of WM/LPL. Staging CT of the neck /chest and abdomen indicated a mild splenomegaly but no lymphadenopathy. Using the International Prognostic Scoring for WM a 5-year-survival of 36% was estimated. A combination chemotherapy (CHOP) was started, and completed further with anti-CD20 rituximab, which resulted in a beneficial overall clinical response. In 2009 small slightly brownish papules appeared on the extensor surfaces of her legs. Upon biopsy extensive dermal deposition of IgM paraprotein diagnostic for cutaneous macroglobulinosis was found. Conclusions: According to current literature only few WM-cases with (both) specific skin lesions have been published so far. The patient is now 78, and still stable. Disclosure: No conflict of interest disclosed. P476

Somatostatin and CXCR4 chemokine receptor expression in lymphoma of mucosa-associated lymphoid tissue (MALT) type Stollberg S.1, Kämmerer D.2, Specht E.1, Schulz S.1, Simonitsch-Klupp I.3, Kiesewetter B.4, Raderer M.4, Lupp A.1 Universitätsklinikum Jena, Institut für Pharmakologie und Toxikologie, Jena, Germany, 2Zentralklinik Bad Berka, Klinik für Allgemein- und Viszeralchirurgie, Bad Berka, Germany, 3Universität Wien, Klinisches Institut für Pathologie, Wien, Austria, 4Universität Wien, Universitätsklinik für Innere Medizin I, Wien, Austria 1

malignancies. Whereas an overexpression of the SSTRs is typically seen with well-differentiated neuroendocrine neoplasms, the CXCR4 is considered to be mainly present in highly proliferative tumor entities. In the present study, the SSTR subtype and CXCR4 expression was evaluated in MALT type lymphoma since comprehensive data on these receptors are still lacking for this tumor entity. Methods: Overall, 55 cases of MALT type lymphoma of both gastric (n = 11) and extragastric (n = 44) origin were evaluated for the SSTR subtype 1, 2A, 3, 4, 5 and CXCR4 expression by means of immunohistochemistry using novel monoclonal rabbit anti-human SSTR or CXCR4 antibodies, respectively. The immunohistochemical stainings were evaluated by means of the Immunoreactive Score and correlated to clinical data. Results: Whereas the CXCR4 was detected in 92% of the cases investigated at a high intensity of expression, the different SSTR subtypes were much less frequently present. Also the intensity of expression within the SSTR-positive tumor samples was much lower as compared to the CXCR4-positive cases. Within the SSTR, the SSTR5 was the most prominent receptor, displaying an expression in about 50% of the cases, followed by the SSTR3, which was present in 35% of the tumors, the SSTR2A, which was detected in 27% of the samples, the SSTR4, which was found in 18% of the cases and the SSTR1, which was present in 2% of the tumors only. There was a correlation between SSTR5 and SSTR3 or SSTR4 expression, but no association between receptor expression and tumor stage. When comparing the different places of origin, tumors of gastric derivation displayed a significantly higher SSTR3, SSTR4 and SSTR5 expression as compared to those of extragastric origin. No differences were seen between gastric and extragastric tumors with respect to SSTR1, SSTR2A and CXCR4 expression. Conclusions: Due to the high expression rate, the CXCR4 may serve as a promising target for diagnostics and pharmacological or radiopharmaceutical therapy of MALT type lymphoma, while the SSTRs appear to be less suitable in this respect. Since with regard to the SSTR only the SSTR5 (and the SSTR3) are expressed in sufficient amount in these tumors, in case of SSTR-based diagnostics and treatment pan-somatostatin analogs should be preferred over octreotide (and derivatives). Disclosure: No conflict of interest disclosed. P477

Bridging and maintenance concepts in systemic anaplastic large cell lymphoma (ALCL) alk+; a single case experience with anti-CD30 therapy followed by ALK inhibition + DLI Hopfer O.1, Kiehl M.1 Klinikum Frankfurt/Oder; I. Medizinische Klinik, Frankfurt/Oder, Germany

1

We report on the case of a young patient with r/r sALCL that was bridged to allogeneic stem cell transplantation with anti-CD30 targeted therapy to control extensive chemotherapy refractory disease. After reaching a very good partial remission conditioning with Cy + TBI was performed before transplantation of a fully matched graft from his sibling. Passing a short interval after complete hematologic recovery and showing no signs of GvHD the patient suffered a severe relapse that was resistant to further anti-CD30 targeted treatment. A salvage strategy was applied using a combined approach with targeted ALK-inhibition plus DLI. This resulted initially in a severe subacute GvHD that could be controlled and transformed into a chronic °I skin GvHD. More, a complete control of his aggressive sALCL was accomplished lasting now more than one year. This case highlights the need for innovative maintenance concepts post allotransplant in aggressive lymphoma patients to support the GvL effect, particularly during the early phase of immune reconstitution. Disclosure: No conflict of interest disclosed.

Introduction: The different somatostatin receptor (SSTR) subtypes and the chemokine receptor CXCR4 are expressed in a wide variety of human

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

143


CONTENTS AUTHOR INDEX

P478

Lymphocyte enhancer binding factor 1 (LEF-1) affects the interaction of Hodgkin lymphoma with endothelial cells Harenberg M.1, Linke F.1, Wilting J.2, Szczepanowski M.3, Klapper W.3, Trümper L.1, Kube D.1 University Medical Centre of the Georg-August-University of Göttingen, Department of Hematology & Medical Oncology, Göttingen, Germany, 2 University Medical Centre of the Georg-August-University of Göttingen, Department of Anatomy and Cell Biology, Göttingen, Germany, 3ChristianAlbrechts-University and University Hospital Schleswig-Holstein, Department of Pathology, Kiel, Germany 1

Introduction: Lymphocyte enhancer binding factor 1 (LEF-1) is a member of the LEF/TCF family of transcription factors central to the canonical Wnt-signaling pathway. Aberrant LEF-1 expression has already been correlated with hematopoietic transformation e.g. chronic and acute lymphatic leukemia and Burkitt lymphoma suggesting a role of defined Wnt-signaling components in lymphoma-microenvironment interactions. However, little is known about Wnt and especially LEF-1 in Hodgkin lymphoma (HL). Hodgkin and Reed/Sternberg (HRS) cells are usually rare in the lymphoma tissue and interactions with the microenvironment are of critical importance for HL pathophysiology. Despite the identification of a complex network of deregulated pathways in HL their impact onto the lymphoma microenvironment has not yet been investigated in detail. To ensure a sufficient nutrient supply the malignant cells have to attract and to interact with endothelial cells of blood or lymphatic vessels but the underlying signaling mechanisms are not yet understood. Methods: HL cell lines with a stable LEF-1 knockdown have been established using lentiviral transfection with small hairpin RNA. For migration assays of Human Umbilical Vein Endothelial Cells (HUVEC) the modified Boyden chamber with collagen I coated membranes and wound healing assays have been used. In vivo chorioallantoic membrane (CAM) assay with chicken eggs was used and evaluated using light microscopy, histology and immunofluorescence staining for blood and lymphatic vessels. Results: LEF-1 expression was found in 4/14 cases of mixed cellularity HL but only in 1/29 nodular sclerosis type HL. Knockdown of LEF-1 in L-428 and KM-H2 cells affects the lymphoma cell secretome. HUVEC migration and wound healing capability is significantly reduced towards supernatant of LEF-1 deficient HL cell lines. This suggests that LEF-1 regulates the expression of factors important for the attraction of endothelial cells. Moreover, in the CAM xenograft model the reduced expression of LEF-1 impairs lymphoma outgrowth, probably as a result of changes in vascularization and/or metabolism. Conclusions: LEF-1 affects the capability of HL cells to attract endothelial cells thereby supporting tumor progression in HL. As consequence LEF-1 might become an interesting target for the treatment of disseminated or relapsed HL cases mainly of a subgroup of the mixed cellularity subtype. Disclosure: No conflict of interest disclosed. P479

Treatment with Horse-Antithymocyte Globulin (hATG) and eculizumab in an aplastic PNH patient: Go or no-go?! Alashkar F.1, Dührsen U.1, Röth A.1 Universitätsklinikum Essen, Hämatologie, Essen, Germany

1

Introduction: PNH therapy is based on supportive or disease modifying strategies. If, in addition, criteria of severe aplastic anemia (SAA) are met, immunosuppressive therapy (IST; hATG/CSA) or bone marrow transplantation are favored. It is not clear if eculizumab has to be discontinued before ATG initiation, as a reduced ATG-efficacy due to interference with complement-mediated cell lysis, unknown pharmacological interference or side effects may result. Case History/Methods: In 2004 a 46-year-old female was diagnosed with PNH (LDH 1865U/l). Due to right popliteal-femoral venous thrombosis 2005 she was put on warfarin and eculizumab was initiated (SHEP-

144

Oncol Res Treat 2015;38(suppl 5):1–270

HERD-Trial), resulting in a significant reduction of hemolytic activity (LDH 251U/l) and transfusion independence. In Jan. 2014 a progressive thrombocytopenia (35/nL) over a 6 month period was seen, despite stable hematologic values: Hb 9.7g/dL, LDH 207U/L, Retics 130.2/nL, WBC 3.7/ nL. Bone marrow diagnostic ruled out progression into AA or MDS. Due to suspicion of immune-mediated thrombocytopenia corticosteroids were started, with no effects. In Mar. 2014 she developed progressive neutropenia (0.8/nL) and became RBC transfusion-dependent with no increased hemolytic activity (LDH 203U/L), confirming SAA in May 2014 (ANC 0,2/nL, PLT 15/nL, Retics 42/nL). Results: In May 2014 hATG (40 mg/kg BW d1–4), CSA (5 mg/kg BW) and prednisone were started. Eculizumab was continued due to the presence of a significant PNH red blood cell population (RBC CD58neg. 30.6%; Retics CD59neg. 97.4%; ARC 170.4/nL) with the risk of (breakthrough) hemolysis and thromboembolic events. After IST a T-cell depletion (0,03/ nL) was seen, similar to non eculizumab treated patients, with hematologic restoration starting by day 5. Following discharge, eculizumab was continued and prednisone was tapered down until day 29. Partial remission was evident by day 147 (ANC 0,8/nL, Hb 8,9 g/dL, PLT 122/nL). Conclusion: In SAA hATG and CSA is a standard IST, resulting in hematologic response in up to 70–80% of pts. This is the third report next to Marotta et al. and Asano et al. 2014, demonstrating an efficient T-cell depletion and clinical response after (h)ATG in pts chronically treated with eculizumab. Further, T-cell depletion does not seem to be altered and no pharmacologic interference or adverse events were seen, proofing a concomitant approach due to significant proportion of PNH RBC a reasonable treatment option in PNH after transition into SAA. Disclosure: Ferras Alashkar: No conflict of interest disclosed. Alexander Röth: Advisory Role: Alexion; Financing of Scientific Research: Alexion; Expert Testimony: Alexion. P480

Epidemiologic overview of congenital dyserythropoietic anemia – updated data from the German CDA registry Theis F.1, Leichtle R.1, Holzwarth K.1, Keppler U.1, Döhner H.1, Bommer M.1,2 Universitätsklinik Ulm, Klinik für Innere Medizin III, Ulm, Germany, 2AlbFils-Kliniken, Hämatologie, Onkologie, Infektiologie und Palliativmedizin, Göppingen, Germany 1

Congenital dyserythropoietic anemia is a very rare hereditary disorder, leading to ineffective hematopoiesis, anemia and secondary hemochromatosis. According to the classification of Heimpel and Wendt (1968) at least four subtypes (CDA I–III, CDA variant) can be distinguished. Diagnosis is based upon morphological examination of blood and bone marrow. Genetic testing can confirm the diagnosis in many cases: CDA I – CDAN1, C15ORF41; CDA II–SEC23B; CDA III–KIF23; variant–KLF 1, GATA-1. Familial cases are autosomal recessive (CDA I, CDA II), autosomal dominant (CDA III) or variable (CDA variant). The German CDA registry was initiated in 1990 by Heimpel and coworkers, and includes cases from all over the world. All patients known to the study group since 1967 have been documented. Data analysis is based upon an ACCESS database. Diagnosis was confirmed using the criteria published previously by Heimpel and Anselstetter (2003). From a minority of patients results from genetic testing are available. We analyzed the prevalence of CDA-subtypes in different continents. CDA IV is summarized within the CDA variant group. At the reporting date (31.12.2013) a total of 873 alive cases from 776 families were registered. Additionally 35 cases are reported to be dead. Geographical analysis shows 715 patients are registered in Europe, 56 in northern America, 13 in southern America, 12 in Oceania (Australia, New Zeeland), 7 in Africa and 70 in Asia. The most frequent subtype is CDA II, followed by CDA I. Most cases are reported in Italy, Spain, Great-Britain, Germany and Israel (data not shown). Detailed information is given in table 1. Prevalence of congenital dyserythropoietic anemia in the different countries is very heterogeneous, reflecting the rarity as well as the lack of

Abstracts


CONTENTS AUTHOR INDEX

awareness of the disease. Nevertheless the diagnosis is under reported due to numerous cases with only mild symptoms misdiagnosed as myelodysplastic syndrome or unclassified hemolytic anemia. Tab. 1. Epidemiology of CDA

Region

CDA I

CDA II

CDA III

Variant

Total

Europa (plus Israel)

154

429

32

100

715

Northern America

10

29

7

10

56

Southern America

0

8

5

0

13

Oceania

4

8

0

0

12

Asia

37

25

3

5

70

Africa

2

4

0

1

7

207

503

47

116

873

Disclosure: No conflict of interest disclosed. P481

Late onset congenital thrombotic thrombocytopenic purpura (TTP) as a cause of relapsing cryptogenic cerebral ischemic events in two adult siblings Schmid K.1, Bommer M.1,2 Alb-Fils-Kliniken, Hämatologie, Onkologie, Infektiologie und Palliativmedizin, Göppingen, Germany, 2Universitätsklinik Ulm, Klinik für Innere Medizin III, Ulm, Germany 1

Thrombotic thrombocytopenic purpura (TTP) is a rare disease characterized by thrombotic events, thrombocytopenia and hemolytic anemia. It is caused by reduced activity of von Willebrand factor cleavage protease (vWFcP, ADAMTS13) in more than 90% of all cases due to acquired antibodies against ADAMTS13. In very rare cases inherited deficiency may be responsible for the clinical appearance, most of the times manifested during childhood or adolescence. Currently, more than 90 different mutations have been identified up to now. We report about a 60 year old female patient with a history of thromboembolic cerebral events and renal failure for more than 20 years. The onset of the disease was in her third decade of life. She suffered from acute renal failure requiring hemodialysis, severe anemia and thrombocytopenia associated with splenic rupture resulting in splenectomy. This episode was classified as hemolytic uremic syndrome (HUS). During the following years hemodialysis could be omitted, but she had several strokes, which resolved without severe permanent neurological damage. She was diagnosed to have cerebral vasculitis and was treated with corticosteroids and cyclophosphamide. During the last event thrombocytopenia, elevated LDH, anemia and low haptoglobin triggered further workup. Finally mutation screening showed both a homozygous ADAMTS13 mutation (c.3178C>T, p.Arg1060Trp) and a MCP mutation (c.1058C>T; p.Ala353Val) in Exon 11. The diagnosis of Upshaw-Schulmann syndrome was established. Family history revealed, that here brother suffered from stroke at the age of 23 and had relapsing cerebral ischemic events during the past 20 years. He was placed in a psychiatric facility because he developed progressive dementia. We also performed hematologic workup in that patient and detected low levels of ADAMTS13 (9%). By doing mutation analysis, we confirmed the same inborn ADAMTS13 defect. This case report of a rare cause of recurrent microangiopathic events in a sibling pair illustrates the need for careful history taking. The detected mutation of ADAMTS13 is reported in the literature to be responsible in patients for adult-onset TTP.

P482

Zeb2 is essential for terminal megakaryocyte differentiation and platelet formation Dobrosch L.1, Li J.1, Riedt T.1, Gütgemann I.1, Huylebroeck D.2, Brossart P.1, Janzen V.1 University Hospital Bonn (UKB), Bonn, Germany, 2Katholieke Universiteit Leuven, Leuven, Belgium 1

Introduction: Zeb2 is a member of the zinc-finger E-box-binding (ZEB) family of transcription factors which has been predominantly associated with the epithelial to mesenchymal transition (EMT) during embryonic development. However, very little is known about the physiological role of Zeb2 in the regulation of homeostasis of adult tissue and especially in the hematopoietic system. Methods: We used the the Mx1-Cre based inducible Zeb2 knockout model to examine the impact in adult murine hematopoieses in a “loss of function” approach. Analyses of peripheral blood trombocyte counts and bone marrow analyses were performed eight weeks following the induction of Zeb2 deletion. In addition we performed ex-vivo functional assay using CFU-MK and liquid culture conditions to test the differentiation ability of the megakaryocytes. Results: Deletion of ZEB2 in the hematopoietic system led to a striking decrease in thrombocyte number associated with an incresed platelet volume compared to the wild type controls. Histological examination of the bone marrow cavity showed significantly more megacaryocytes in the of Zeb2Δ/Δ Mx1-Cre animals; however these cells displayed an abnormall morphology with drastically reduced cytoplasm and often found to cluster in small groups. As Zeb2Δ/Δ Mx1-Cre mice developed extramedular hematopoiesis we also observed a massiv infiltration of the spleen with similar atypical megacaryocytes. CFU-MK assays revealed no mature megakaryocytic colonies determined by acythylcholinesterase staining, indicating a differentiation defect in absence of Zeb2. FACS-analysis of cultured megacaryocytes did not show perturbation in formation of polyploid cells, as there was no reduction in DNA replication. Thus, we confirm that megakaryocytes still can be produced in-vitro but show differentiation defects at the terminal stages of maturation. Conclusions: Zeb2 ablation in the hematopoietic compartment of the adult mouse led to significant alterations of the megacaryopoesis and thrombopoiesis. These changes are at least in part due to a maturation impairment. Disclosure: No conflict of interest disclosed.

Posterdiskussion Querschnittsthemen I P483

Evaluation of Vulnerable Elders Survey, G8 questionnaire and Predictors of Toxicity as screening tools for frailty and toxicities in geriatric patients with cancer Hentschel L.1, Rentsch A.1, Hornemann B.1, Lenz F.2, Baumann M.1,3, Ehninger G.1,2, Schmitt J.4, Schuler M.K.2,5 Universitätsklinikum Carl Gustav Carus, Universitäts KrebsCentrum, Dresden, Germany, 2Universitätsklinikum Carl Gustav Carus, Medizinische Klinik I, Dresden, Germany, 3Universitätsklinikum Carl Gustav Carus, Klinik für Strahlentherapie und Radioonkologie, Dresden, Germany, 4Universitätsklinikum Carl Gustav Carus, Zentrum für Evidenzbasierte Gesundheitsversorgung, Dresden, Germany, 5Helios-Klinikum Emil von Behring, Klinik für Innere Medizin II, Berlin, Germany 1

Disclosure: Kathrin Schmid: No conflict of interest disclosed. Martin Bommer: Financing of Scientific Research: Vortragshonorare der Firma Alexion.

Introduction: A comprehensive geriatric assessment (CGA) can help to improve cancer care in elderly patients. As completing a full CGA is both time and resource consuming, screening instruments such as the Vulnerable Elders Survey (VES-13), the G8 questionnaire (G8) or the Predictors of Toxicity (POT) are available for predicting frailty and toxicities. For the first time, we tested the predictive qualities of all three instruments com-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

145


CONTENTS AUTHOR INDEX

pared to a full CGA during routine admission process at a Comprehensive Cancer Center (CCC). Methods: 84 patients with a first appearance at a CCC were included in this cross-sectional, cohort study. As part of the routine admission procedure, patients conducted a CGA, consisting of six dimensions (cognition, mood, instrumental activities of daily living, nutritional status, polypharmacy, falls) and the VES-13, G8, and POT. Analysis was performed for sensitivity, specificity, positive predictive value (ppv), and for the negative predictive value (npv). The area under the curve (AUC) of instruments’ ROC-curves was calculated. Results: 84 patients (52 men) with a median age of 73 years (range: 63– 93) participated. 30 Patients (35.7%) were classified as “frail” by the CGA. There was no significant difference for ECOG status or age between frail and non-frail patients. Predictive values and AUC of the screening instruments are reported in Table 1. Tab. 1. predictive values and AUC

VES13

G8

95% confidence interval

95% confidence interval

95% confidence interval

Sensitivity

57,1%

44,5– 69,8%

38,3%

25,9– 50,7%

72,7%

61,2– 84,3%

Specificity

79,2%

68,8– 89,5%

62,5%

50,1– 74,9%

65,2%

52,9– 77,6%

positive predictive value

61,2%

48,7– 73,6%

37,0%

24,6– 49,3%

54,6%

41,6– 67,5%

negative predictive value

76,3%

65,4– 87,1%

63,8%

51,6– 76,1%

80,6%

70,4– 90,9%

Area under the Curve

0.732

0.303

Methods: From 01–10 to 31–12–2012 medical staff was asked to answer the SQ for each cancer patient on their daily appointment-list of pts. In case of the answer: “No, I would not be surprised”, physicians were further asked to indicate if the expected survival time could even be less than 3 months. All pts visiting the outpatient clinic were analyzed by age, gender, disease, stage of disease, current therapy, and advance care planning. Patients were reassessed as to survival after 18 months. Results: The SQ was answered for 651 of 687 pts (94.7%). 8 pts were lost for follow-up. The SQ was answered with “No, I would not be surprised” for 139 (21.6%) pts, of whom 37 were expected to live less than 3 months (5.8% of all pts). After 12 months, 71 pts (51%) of the “No, I would not be surprised” group had died, the corresponding number in the “Yes, I would be surprised” group was 37 (7.3%). Of the 37 pts with an estimated life expectancy of less than 3 months, 17 (46%) died within this time frame, another 12 (32%) during the following 9 months. The hazard ratio to die within 12 months was 5.2 when the SQ was answered with “No, I would not be surprised” (Fig. 1). Conclusions: The results of our study are in line with previous studies showing the utility of the SQ to identify patients with a greatly increased risk of 1-year mortality. The SQ is a feasible and intuitive tool. Answering the SQ with “No” should rise the treating physician’s awareness that his/ her patient has a significant probability to die within one year, so that issues beyond active cancer treatment should be addressed as well.

0.781

Conclusions: The predictive values of three widely used screening instruments for frailty were assessed. 36% of all patients were classified as frail. Except for sensitivity of the G8, other values were in the range of published literature. The frailty in those studies was higher (range 67–82%). With a high sensitivity (72.7%) and a npv (80.6%), the POT might be the overall most promising instrument for routine use in a CCC. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

P484

“Would I be surprised if this patient died in the next year?” – Prognostic significance of the “Surprise” Question in a university hematology and oncology outpatients clinic Halbe L.1,2, Gerlach C.1,2, Hess G.2,3, Wehler T.2,3, Theobald M.2,3, Weber M.1,2 University Medical Center of the Johannes Gutenberg-University of Mainz, Interdisciplinary Palliative Care Unit, III. Department of Medicine, Mainz, Germany, 2University Medical Center of the Johannes Gutenberg-University of Mainz, University Cancer Center (UCT Mainz), Mainz, Germany, 3University Medical Center of the Johannes Gutenberg-University of Mainz, III. Department of Medicine, Mainz, Germany 1

Introduction: Timely identification of patients in need for palliative care remains a major challenge. The ‘Surprise’-Question SQ (“Would I be surprised if this patient died in the next year?”) has been shown to be a simple, feasible, and effective tool to identify patients with a greatly increased risk of 1-year mortality (Moss AH et al. J Palliat Med. 2010). In the context of introducing early palliative care into our service, we tested the SQ in our hematology and oncology outpatient clinic. Here we report about first results concerning the prognostic significance of the SQ.

146

Fig. 1. Survival Curves of “yes” and “no” response.

Oncol Res Treat 2015;38(suppl 5):1–270

P485

Low incidence of cytostatics extravasation and surface contamination through good risk management Guger-Halper U.1, Stangl T.A.2, Zapfel S.3, Stangl W.M.4 KH Oberwart, Anstaltsapotheke,Zentrale Zytostatika-Aufbereitung, Oberwart, Austria, 2VU University, Amsterdam, Netherlands, 3KH Oberwart, Interne Abteilung, Oberwart, Austria, 4KH Oberwart, Hämato-/Onkologie und Palliativmedizin, Oberwart, Austria 1

Introduction: Extravasation of cytostatics and surface contaminations are emergencies in oncology. The frequency of occurrence of extravasation in cancer patients is reported in the literature with 0.45% to 6.4%. Based on the number of infusions, there is a proportion of 0.01% to a maximum of 0.9%. Extravasation of cytostatics are not documented in many cases, and are also often not noticed by the clinicians and the patients. Methods: We have prospectively documented all cytostatics incidents on our oncology unit, involving a standardized incident report. In addition to the patient data, the type, concentration and mode of administration

Abstracts


CONTENTS AUTHOR INDEX

of the cytostatic drug were cited. The puncture site, the venous access, the local symptoms and size of the lesion and the measures taken, a time stamp and risk factors for wound healing have been documented.Every two months all complications were discussed by all participants in a separate conference in detail.It was differentiated between surface contamination and extravasation into the subcutis. Results: During the period of two years (2013 and 2014) from 5489 cytostatic infusions, 3 cases (0.05%) of extravasation were detected in total. Panitumumab, Irinotecan and 5-FU were involved. In each case there was skin swelling above the port. In the case of irinotecan burning at the skin-swelling occurred too.Tissue damage did not occur in any case. The time from start of the infusiont to perception by a patient or clinician averaged 8.6 minutes (5–15).The estimated infusion volume was 6 ml (3– 10). In the case of Irinotecanparavasation a not measurable amount was aspirated. In addition, there were 8 cases (0.15%) from surface contamination.These were supplied in accordance with the departmental guideline. There was neither a patient damage nor a harm of the staff. Conclusions: We found a low incidence of cytostatic incidents. No case of extravasation resulted in permanent damage at the injection site. In addition to patient education, the annual training of the care provider and the bi-monthly complication- conference were the main factors in order to avoid permanent damage to the patients and staff at our Oncology Unit. Disclosure: No conflict of interest disclosed. P486

Next generation chemotherapy (CTx) management software system to ensure and increase patient safety Szymaniak-Vits M.1,2, Klug J.3, Reinhardt H.1, Kaiser S.1,2, Otte P.1,2, Hug M.4, Claussen A.5, Duyster J.1,2, Engelhardt M.1, Ruch M.3 University Medical Center Freiburg, Department of Hematology and Oncology, Freiburg, Germany, 2Comprehensive Cancer Center Freiburg (CCCF), Freiburg, Germany, 3MPS – Medizinische Planungssysteme GmbH, Freiburg, Germany, 4 University Medical Center Freiburg, Pharmacy, Freiburg, Germany, 5University Medical Center Freiburg, Central Quality Management, Freiburg, Germany 1

Introduction: The risk of medication errors in complex CTx regimen is high (Walsh, JCO 2008). In order to increase patient safety, the Dept. of Hematology & Oncology, University Medical Center Freiburg (UMF) approached this challenge by using a CTx management software system (CTxMS). Combined with comprehensive monitoring, consisting of manual checks done by a quality management team (QMT), it was possible to achieve significant reduction in medication errors (Markert, ... Engelhardt, IJC 2009). We here describe the process of advancing this tool into a next generation CTxMS, designed to provide universal access to similar results. Methods & Results: The original CTxMS includes detailed CTx protocols, complete with comedication, SOPs & clinical pathways, which allows a reduction of potentially lethal medication errors by at least 47%. Complementary to checks done by the clinical pharmacy and the medical staff, specific monitoring processes, including an SAE management process, were followed by a trained QMT and intercepted virtually all observable errors. In a project funded by the Federal Ministry of Education & Research (BMBF) numerous of these effective, yet manually executed, plausibility and correctness checks were integrated into a novel software for a more efficient process and a further reduction of manual errors. Via software interfaces, the CTxMS was connected to the existing IT infrastructure to allow a seamless integration in up- and downstream processes. This leads to a speedup of the complete process and additional error reduction (ER) -e.g. a manual transcription of CTx orders into the pharmaceutical software system was no longer necessary. Thus, it is possible to implement a comprehensively documented and revision controlled process. Additionally, the flexibility of the software´s web based architecture allows the simple transfer to other health facilities, in order to enable as many facilities as possible to achieve similar outstanding results in error reduction and increased efficiency.

Abstracts

Conclusions: UMF´s next generation CTxMS uses automated CTx monitoring to allow for superior patient safety and an efficient and revision controlled automation of a formerly manual, comprehensive monitoring process. Its flexible architecture allows a simple and feasible transfer to other facilities, enabling small and larger sized hospitals in particular to achieve comparable process optimizations despite limited financial and technical resources. Disclosure: Magdalena Szymaniak-Vits: No conflict of interest disclosed. Markus Ruch: Expert Testimony: BMBF research project “QmacPro – Qualitätsmanagement für chemotherapeutische Planungsprozesse“ (ID: 13GW0064A). P487

O-PIS.meine akte – my patient record for my physicians Pareigis S.1, Kunde-Krüger J.2, Ebert A.2 SHG Leukämie-u. Lymphompatienten Halle (S.), Schkopau / OT Ermlitz, Germany, 2IT-Consult Halle GmbH, Halle, Germany 1

Introduction: Why do we need a web-based Patient Record for Patients? Background: A patient record can be comprised of many folders depending on the nature of the course of disease. The physical weight alone of carrying such folders, along with the tedious process of searching for relevant information is hardly acceptable. Methods: O-PIS.meine akte is a web-based application. It is accessible from any device (PC or Tablet) with Internet access. The patient digitizes and classifies his or her documents so that they can be centrally stored in O-PIS.meine akte. A physician will receive access credentials before the scheduled appointment with the patient. The physician can then securely access the patient’s record and add relevant documents as part of an ongoing treatment. Functionality: · Secure storage and classification of electronic documents · Efficient searching and information filtering · Chronological anamnesis · Apointment scheduling · Strict access and permission control · Medical documentation · Episode concept · Disease diary · Logging of treatments, diagnosis and findings · Emergency access Result: The O-PIS.meine akte web-based application makes it possible for patients as well as doctors to have fast and secure access to patient records from anywhere in the world. Conclusion: The immediate interdisciplinary availability of reports on anamnesis, patient discharge, CT-MRT-PET data, course of disease, medication as well as ordered treatment is securely provided by means of using the O-PIS.meine akte application. Patients can independently upload documents such as referrals, vaccination cards, allergy IDs, availability and disease diaries. A pilot project is expected to start in the near future. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

147


CONTENTS AUTHOR INDEX

P488

P489

Web-based stress management program (STREAM) for newly diagnosed cancer patients: An ongoing randomized, wait-list controlled intervention study. Usability results

Correlation between somatic comorbidities and anxiety and depression in patients after allogeneic hematopoietic stem cell transplantation

Grossert A.1, Heinz S.2, Berger T.3, Gaab J.4, Alder J.5, Scherrer S.6, Urech C.6, Hess V.1 1

Balsiger C.1, Mumm F.1, Holler E.2, Herzberg P.Y.3, Hilgendorf I.4, Hemmati P.5, von Harsdorf S.6, Greinix H.7, Mitchell S.A.8, Wolff D.2, Heussner P.1

2

1

Universitätsspital Basel, Medizinische Onkologie, Basel, Switzerland, Universität Basel, Psychologische Fakultät, Abt. für Mensch und Maschinen Interatkion, Basel, Switzerland, 3Universität Bern, Klin. Psychologie, Bern, Switzerland, 4Universität Basel, Klin. Psychologie, Basel, Switzerland, 5Universität Basel, Psychologische Fakultät, Basel, Switzerland, 6Universitätsspital Basel, Gynäkologische Sozialmedizin und Psychosomatik, Basel, Switzerland

Introduction: Many cancer patients (pts) suffer from high psychological distress and impaired mental adjustment. Psycho-oncological interventions reduce perceived stress, anxiety and depression and increase quality of life. Internet interventions allow for independence in time and place and overcome many barriers for seeking face-to-face support. The present study aims at investigating the usability, feasibility and preliminary efficacy of the first German web-based stress management intervention (STREAM: STREss Aktiv Mindern) for newly diagnosed cancer pts Methods: 120 newly diagnosed cancer pts will be included in an ongoing randomized controlled wait-list intervention trial. The intervention program consists of 8 weekly modules, based on well-established stress management manuals and adapted to the context of cancer and to the web-format. In a first step, usability of the web-based program has been investigated to identify potential issues in the design of the program. In a second step, feasibility will be assessed with regard to recruitment, pts’ characteristics, compliance and adherence. Preliminary efficacy will be longitudinally assessed with validated questionnaires (DT, HADS, FACIT-F). Results: The RCT started in June 2014: so far 61 pts were screened and 21 included. Pts are from Switzerland, Germany and Austria. Prior to the RCT, usability was assessed in 12 cancer pts (female n = 6, male n = 6; mean age: 61.6, range: 37–77 years) with the system usability scale (SUS). Mean SUS score was 83.6, signifying a high usability (cut-off = 70). The pts described the program as clear and structured. Acceptance of the intervention program among pts (including reliability and professional appearance of the website) was high. Conclusions: Overall usability and satisfaction with the newly developed stress management program was high in the targeted population. On the basis of the usability test the program was optimized in specific areas before starting with the randomized controlled wait-list intervention trial. Results of the RCT will provide a rationale for future RCTs assessing the efficacy of STREAM in a more homogenous patient population. The webbased stress management intervention might be an additional support for pts who suffer high levels of stress during cancer treatment. An effective stress management program might have an impact not only on psychological outcome but also on treatment side effects and, ultimately, on the course of disease. Disclosure: No conflict of interest disclosed.

Klinikum Großhadern, Ludwig-Maximilians-Universität, Dept. of Internal Medicine III, München, Germany, 2Universitätsklinikum Regensburg, Dept. of Internal Medicine III, Regensburg, Germany, 3Helmut-Schmidt-Universität / Universität der Bundeswehr Hamburg, Dept. of Personality Psychology and Psychological Assessment, Hamburg, Germany, 4Universitätsklinikum Jena, Dept. of Internal Medicine II, Jena, Germany, 5Charité – Universitätsmedizin Berlin, Dept. of Hematology and Oncology, Berlin, Germany, 6 Universitätsklinikum Ulm, Dept. of Internal Medicine III, Ulm, Germany, 7 Medizinische Universität Graz, Dept. of Hematology, Graz, Austria, 8National Cancer Institute, Bethesda, United States

Introduction: Chronic Graft-versus-host disease (cGvHD) is a frequent and serious complication after allogeneic hematopoietic stem cell transplantation (HSCT) with effects on quality of life, morbidity and mortality. In addition to cGvHD itself, coexisting somatic comorbidities may contribute to anxiety and depression and thereby affect the psychological well-being of patients after HSCT. We therefore investigated the correlation of somatic comorbidities with anxiety and depression in patients with and without cGvHD. Methods: In this international multicenter trial, we studied a cohort of 208 patients (male: n = 106; female: n = 102) who had received HSCT. Patients were evaluated prospectively using the Hospital Anxiety and Depression Scale (HADS). Data for the Post-Transplant Multimorbidity Index (PTMI) were collected retrospectively. The investigated comorbidities of interest were hypertension (compensated and uncompensated), diabetes mellitus, infection (mild, moderate and severe), osteopenia/osteoporosis, hypothyroidism and underweight. Results: In our cohort, 137 patients had cGvHD while 71 patients had no cGvHD. Among patients with cGvHD, we identified multiple somatic comorbidities that were associated with depression. Patients with compensated hypertension on treatment more frequently had depression (35.3%) compared to patients without hypertension (9.2%, p < 0.01). Similarly, patients with diabetes mellitus requiring treatment were significantly more likely to have depression (50.0%) than patients without diabetes (12.5%, p < 0.01). Among patients with osteopenia/osteoporosis (T-score ≤ -1.5 or on treatment with a bisphosphonate), 32.0% had depression compared to 12.1% of patients without osteoporosis (p < 0.05). Among patients without cGvHD, mild infections and underweight were significantly (p < 0.05 and p < 0.01, respectively) associated with anxiety. In contrast, we found no association between moderate/severe infections and hypothyroidism and anxiety or depression in either of the two patient subgroups. Conclusions: Morbidity after HSCT is triggered multifactorially. Beside the influence of cGvHD itself, we identified several somatic comorbidities that were associated with anxiety and depression. Further investigation with prospective trials is needed in the field of cGvHD, comorbidities and quality of life. Disclosure: No conflict of interest disclosed. P490

Psychosocial status of acute and former childhood cancer patients searching for work in Austria – an evaluation study Nagele E.1, Fürschuß C.1, Wiegele K.1, Mohapp A.1, Urban C.1 Univ.-Klinik für Kinder- und Jugendheilkunde; Medizinische Universität Graz, Abteilung für pädiatrische Hämato-Onkologie, Graz, Austria 1

Introduction: Quality of life and steps towards planning a career and choosing a profession can be negatively influenced in the adolescence of cancer patients.

148

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

The current study was performed to evaluate related influencing factors for childhood cancer patients actively searching for work and participating in the Austrian project “Jugend und Zukunft” (“Youth and Future”) supported by “die Berater®”, a consultancy focusing on training and development. Methods: Questionnaires including psychological influencing factors, resources and possible aspects of distress were forwarded to patients, their parents and coaches, up to 9 months via an Online-Tool. Results: Up to now 5 participants including two patients still in rehabilitation, their parents and one coach, were analysed. Patients were older than 18 years and had finished apprenticeship or were educated on a post-secondary level. Ewing`s sarcoma or a brain tumour was diagnosed with 13 years respectively 8 years. Both were willing to work and were searching for 6 and 23 months. They indicated to have a high quality of life. Self-esteem and satisfaction with life were assessed differently, but rather high. Parents rated children’s strengths and limitations as realistic, but self-esteem as elevated. Coaches replied that aiming for autonomy and empowerment are also important for their clients next to find work. Conclusions: For an optimal psychosocial care, measures for the improvement in quality should be adapted or implemented based on the results. Disclosure: No conflict of interest disclosed. P491

Effects of an inpatient rehabilitation program on physical performance and fatigue of patients after allogeneic stem cell transplantation Lotze C.1, Krtschil M.1, Mehnert K.1, Wollina K.1, Korfee S.1, ErdmannReusch B.1 Klinik Bavaria, Kreischa, Germany

1

Introduction: The impairment of physical performance and psychological status are frequent and relevant problems of patients after allogeneic stem cell transplantation. Several studies suggests that physical exercise therapy may help to improve physical capacity and to reduce fatigue. Methods: 131 patients undergoing an allogeneic stem cell transplantation participated at the study. The patients carried out an endurance and resistance training 5–6 times weekly for three weeks. Training consisted of walking on a treadmill or biking on a stationary bike at 80% ± 5% of maximal heart rate. Resistance training consisted of several exercise sets on 4–6 different devices to improve e.g. limb strength. Physical functioning has been evaluated through the Six-Minute Walk Test (6MWT), physical working capacity (PWC) on an ergometric bike and the voluntary isometric maximal contraction strength of the quadriceps cruralis and the performance status (Karnofsky-Index) at the beginning and at the end of the study. The psychological status has been evaluated using the Fatigue-Scores (LASA, MFI), Hospital Anxiety and Depression Scale (HADS-D and HADS-A). We are examined the heart rate variability and its correlation with these parameter. Results: The exercise program resulted in clinical and statistical relevant improvement of physical and psychological performance. We observed a significant reduction of fatigue scores (25%) and improvement the performance status, 6MWT, PWC and muscle strength. The heart rate variability was correlated with physical performance status, fatigue scores and 6MWT. Conclusions: A structured inpatient exercise program is feasible and helps to increase physical performance and to reduce fatigue of patients after allogeneic stem cell transplantation. Disclosure: No conflict of interest disclosed.

P492

Early electronic screening for psychological distress and need for psychooncological support in a Comprehensive Cancer Center Hentschel L.1, Hornemann B.1, Trautmann F.2, Schmitt J.2, Schuler M.3,4 Universitätsklinikum Carl Gustav Carus, Universitäts KrebsCentrum, Dresden, Germany, 2Universitätsklinikum Carl Gustav Carus, Zentrum für Evidenzbasierte Gesundheitsversorgung, Dresden, Germany, 3Universitätsklinikum Carl Gustav Carus, Medizinische Klinik I, Dresden, Germany, 4Helios-Klinikum Emil von Behring, Klinik für Innere Medizin II, Berlin, Germany 1

Introduction: Almost a third of cancer patients suffer from comorbid psychological disorders. The proportion of patients experiencing clinical important distress is even higher. National guidelines emphasize the need to screen for psychological distress in order to implement early psychooncological care. Different screening instruments are used throughout Germany, including the NCCN-Distress Thermometer (DT) and the Hornheider Screening Inventory (HSI). At our Comprehensive Cancer Center (CCC) patient reported outcomes are assessed electronically and made available immediately to treating physicians via the clinic information system. Methods: As part of the routine admission procedure, data of 126 consecutive patients were assessed between May and October 2014. Distress was measured using the DT, a single-item analogue scale ranging from 0 (no distress) towards 10 (highest possible distress). Need for psychooncological support was measured using the HSI, a 7-item questionnaire, ranging from 0 (no need) to 14 (severe need). Cut-offs were set to ≥5 points for the DT and ≥4 for the HSI according to respective guidelines. Descriptive analysis was conducted for the total number of patients exceeding these cut offs. Furthermore, we assessed the number of patients classified as distressed but not in need for psychooncological support and vice versa. Correlation between scores in DT and HSI was calculated. Results: Most patients were male (66.7%), average age was 63 years and 80 patients (63.5%) were suffering from gastrointestinal tumours. Overall, the item on HSI and DT was completed by 125 (99.2%) and 108 (85.7%) patients and analysis was conducted for those 108 patients with both scores. 67 persons (62.0%) were assessed as distressed by the DT, 55 patients (50.9%) were in need for psychooncological support. 22 (20.4%) patients were screened as distressed but indicated no need for psychooncological support. 10 (9.3%) patients reported need for psychooncological support but were not screened as distressed. There was a moderate positive correlation (Spearman’s rho = 0.59) between scores of DT and HSI (p < 0.001). Conclusions: Early electronic screening reveals a high level of psychological distress and need for psychooncological support in a significant amount of patients. An open question remains whether those distressed patients indicating no need for professional assistance handle the affective burden themselves and how their distresses changes longitudinal. Disclosure: No conflict of interest disclosed. P493

Immunotherapeutic strategies for soft-tissue sarcoma: Preclinical results of Natural Killer cell augmentation by ex vivo expansion and use of an anti-GD2 antibody Bücklein V.1,2, Jorg T.2, Pass D.2, Krupka C.2, Schlegel P.3, Lang P.3, Hoffmeister sen. H.4, Lindner L.1, Hiddemann W.1, Subklewe M.1,2 Klinikum der Universität München, Medizinische Klinik und Poliklinik III, München, Germany, 2Helmholtz Zentrum, München, Germany, 3 Universitätsklinikum Tübingen, Abteilung für Kinderheilkunde I, Tübingen, Germany, 4Zellwerk GmbH, Oberkrämer, Germany 1

Introduction: Patients with advanced soft-tissue sarcoma (STS) have a poor prognosis with high relapse rates after multimodal therapy. Immunotherapeutic strategies aim to eradicate quiescent malignant cells as possible origin of relapse. Natural Killer (NK) cells are targets of immunotherapeutic interventions because tumor-induced immunotolerance has

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

149


CONTENTS AUTHOR INDEX

been associated with dysfunctional NK cells. To restore the lytic activity of NK cells, ex-vivo expansion or use of monoclonal antibodies directed against cancer-specific antigens are promising strategies. We compared the cytotoxic activity of NK cells of STS patients and healthy donors. Augmentation of cytotoxicity was assessed after long-term expansion in a bioreactor with Interleukin-2-containing medium. Additionally, the effect of ch14.18, a monoclonal antibody directed against GD2 (an antigen frequently expressed on STS subentities) on NK cell mediated cytotoxicity was tested. Methods: Cytotoxicity was measured in a standard 51Cr release assay. NK cells were tested for their lytic capacity before and after expansion in a long-term bioreactor system. Changes in the expression pattern of activating NK cell receptors before and after expansion were analyzed by flow cytometry. Additionally, the additive effect on cytotoxicity of ch14.18 was evaluated. Results: Compared to NK cells of healthy donors, patient NK cells showed impaired cytotoxicity against K562, a standard NK cell target cell line. In contrast, lytic activity against STS cell lines of NK cells of both patients and healthy donors was comparably low. Expansion of NK cells of patients (n = 4) and healthy donors (n = 3) resulted in cell numbers of up to 90-fold of baseline cell counts, and led to a significant increase (of up to 25-fold) in cytotoxicity against K562 and STS cell lines. Expression of activating NK cell receptors (e.g. NKG2D) was significantly upregulated on expanded NK cells. Addition of ch14.18 caused an increase in cytotoxicity of unstimulated NK cells of healthy donors and patients against GD2-expressing STS cell lines (SW872 and TC-71) of up to 7- and 1.4-fold, respectively. For expanded NK cells of healthy donors, no additional effect of ch14.18 was seen. Conclusions: NK cells of STS patients show impaired cytotoxic activity. Long-term expansion and use of anti-GD2 antibodies are promising approaches to restore their lytic capacity. Disclosure: No conflict of interest disclosed.

Posterdiskussion Sonstige Themen I P494

LEONIS- Long term everolimus observation non-interventional study in pancreatic neuroendocrine tumors (pNET) Wiedenmann B.1, Stauch M.2, Akca A.3, Kleylein-Sohn J.4, Klausmann M.5, Tessen H.-W.6 Charité – Universitätsmedizin Berlin, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Berlin, Germany, 2Praxis für Hämatologie und Onkologie, Kronach, Germany, 3Lukaskrankenhaus Neuss, Visceral and endocrine Surgery, Neuss, Germany, 4Novartis Pharma GmbH, Nürnberg, Germany, 5Praxis für Hämatologie, Onkologie, Diabetologie und Gastroenterologie, Aschaffenburg, Germany, 6Praxis für Onkologie, Goslar, Germany 1

Introduction: The aim of this prospective multicentric non-interventional study is to collect real-world data on the treatment of pNET patients with everolimus focusing on compliance, QoL and pharmacoeconomics. Moreover, data on safety and efficacy of everolimus treatment are obtained. Methods: For each patient clinical data are documented by the investigator every three months following routine care over a maximum observation period of 12 months. Results: Here, we present first results on baseline characteristics and treatment effectiveness from an ongoing final analysis. 62 patients were included in the full analysis set (FAS). The FAS was subdivided into FASp (pretreated with everolimus, n = 20) and FASs (everolimus-naïve, n = 42). While 24 patients (39%) completed 12 months of documentation, 38 patients (61%) discontinued prematurely, mainly due to disease progression or death (n = 21, 34%). The Ki-67 index had been determined in 75% of patients, of which 22% had Ki-67 <2%, 33% had Ki-67 2–4% , 20% had

150

Oncol Res Treat 2015;38(suppl 5):1–270

Ki-67 5–9% , 24% had Ki-67 10–20%. Metastases were found in 95% of patients. 61% of patients had a hepatic tumor load of >10%. 42% of patients received surgery prior to everolimus therapy and 69% received one or more prior antineoplastic therapies including Somatostatin analoga (SSAs) (44%), Streptozotocin (21%), Sunitinib (13%), PRRT (19%), TAE/ TACE/SIRT/RFA (8%). The median treatment duration in the study was 6.6, 6.3 and 7.8 months for FAS, FASs and FASp patients, respectively. In addition, FASp patients had been pretreated with everolimus for a median duration of 3.0 months. The mean dose was 9.3 mg everolimus for FASp and 8.0 mg for FASs, respectively. The median PFS was 8.6 (95% CI 5.3–13.8) months for FAS patients, 11.3 (95% CI 3.9–23.1) months for FASp patients and 7.2 (95% CI 3.9–13.8) months for FASs patients. The overall response rate based on the investigator`s assessment at the end of study was 12%, 5% and 15% in FAS, FASp and FASs patients, respectively – resulting in disease control rates of 57% (FAS), 63% (FASp), 54% (FASs). The overall assessment of compliance was `very good` for 61%, `sufficient` for 29% and `insufficient` for 3% of patients according to investigator judgement. Conclusion: In a real-world setting everolimus treatment was associated with high therapy compliance, a median PFS of 9 months and a disease control rate of 57%. More data on QoL, safety and PFS, are currently processed and will be incorporated into the poster. Disclosure: No conflict of interest disclosed. P495

Tumor infiltrating lymphocytes and PD-1/PD-L1 expression differ between low and high grade neuroendocrine tumors – first results of a protein- and array-based pilot study Spenke C.1,2, Jöhrens K.3, Arsenic R.3, Briest F.1,4, Lammert H.3, Kaemmerer D.5, Hummel M.3, Scheibenbogen C.2, Busse A.6, Grabowski P.1,2 Charité Campus Benjamin Franklin, Medizinische Klinik I, Gastroenterologie, Infektiologie, Rheumatologie, Berlin, Germany, 2Charité-Campus Virchow Klinikum, Institut für Immunologie, Berlin, Germany, 3Charité – Universitätsmedizin Berlin Campus Mitte, Institut für Pathologie, Berlin, Germany, 4Freie Universität Berlin, Institut für Biochemie, Berlin, Germany, 5 Zentralklinik Bad Berka GmbH, Klinik für Viszeralchirurgie, Bad Berka, Germany, 6 Charité Campus Benjamin Franklin, Medizinische Klinik für Hämatologie, Onkologie, Tumorimmunologie, Berlin, Germany 1

Introduction: Although much progress has been made in the past 2–3 years in terms of understanding signaling pathways in gastroenteropancreatic neuroendocrine tumors (GEP-NENs), approved treatment options are still limited. Cancer Immunotherapy has been announced as the breakthrough of the year in 2013 and might become an integral part of the clinical management strategy for solid tumors including GEP-NENs. Therefore we need to know which immune escape mechanisms are relevant in high and low grade NEC/NENs in order to develop immunotherapeutic strategies. Aim(s): We aim to characterize the tumor microenvironment (TME) of NEN/NECs on the cellular level in order to define relevant immunophenotypes. Methods: TMAs of 25 poorly differentiated neuroendocrine carcinomas (PDECs) of all localizations as well as 47 well differentiated ileal neuroendocrine neoplasias (WD-NEN) of two different institutions (Charité Berlin and Zentralklinik Bad Berka GmbH) were stained for CD3 expression; 14 PDECs and 25 WD-NENs were stained for PD-L1 expression. A pilot series of 12 tumors (6 WD-NENs and 6 PDECs was analyzed with the PanCancer® Immune profiling multiplex gene expression analysis Panel (Nanostring® Technologies) for the differential expression of immune-relevant genes. Results: Much more CD3 positive lymphocytes were seen in PDECs than in WD-NENs (50–200/HPF vs. 5–10/HPF). In addition, PD-L1 expression was more frequently observed on tumor cells in PDECs (6/6 primary tumors, 3/3 lymph node metastases, 7/7 distant metastases, 100%) than in WD-NENs (13/22 primary tumors, 7/10 lymph node metastases, 1/3 distant metastases, 60%).The multiplex gene expression analysis of well vs poorly differentiated neuroendocrine tumors revealed 186 significantly

Abstracts


CONTENTS AUTHOR INDEX

differentially expressed genes, e.g. an up-regulation of PDCD1 (encoding for PD-1) in PDECs. Highly significant changes could be found in 12 genes of the MAPK-pathway, the complement system and of chemotaxis (e.g. CXCL10, CXCL11). A complete list of all relevant genes and correlation with clinical data will be shown on the meeting. Conclusion: Our preliminary data indicate that PD1/PD-L1 and further targets of the T-cell mediated immune response may be promising new targets – especially in poorly differentiated NECs, where new therapeutic options are highly needed. Disclosure: No conflict of interest disclosed. P496

Cancer patients’ preferences regarding control in decision making and possible determinants. Findings from a German outpatient cancer center Schuler M.1,2, Schildmann J.3, Trautmann F.4, Hornemann B.5, Hentschel L.5, Ehninger G.2,5, Schmitt J.4 Helios-Klinikum Emil von Behring, Klinik für Innere Medizin II, Berlin, Germany, Universitätsklinikum Carl Gustav Carus der Technischen Universität Dresden, Medizinische Klinik I, Dresden, Germany, 3Ruhr-Universität Bochum, Institut für Medizinische Ethik und Geschichte der Medizin, Bochum, Germany, 4 Universitätsklinikum Carl Gustav Carus der Technischen Universität Dresden, Zentrum für evidenzbasierte Gesundheitsversorgung (ZEGV), Dresden, Germany, 5Universitätsklinikum Carl Gustav Carus der Technischen Universität Dresden, Universitäts KrebsCentrum, Dresden, Germany 1 2

Introduction: Patient centered information and decision making is a requirement from an ethical, legal and also clinical perspective. However, there is scarcity of data on patients’ decisional control preferences (DCP) and their possible clinical determinants elicited during routine clinical care. Methods: Patients presenting for the first time were offered a self-administered electronic questionnaire with questions on quality of life (EORTC QLQ C30), needs for psycho-oncological support (Hornheider Screening Instrument), psychosocial distress (NCCN Distress-Thermometer), decisional control preferences (control preference scale), nutritional assessment (Mini-Nutritional Assessment) and patients estimated performance status. Patient reported outcomes (PROs) were combined with data of patients’ file. Hypothesising that older age, more progressed disease and higher needs for psycho-oncological support are related to leaving the responsibility of the treatment decision with the physician we investigated possible determinants of the patients DCP by applying cross sectional analysis. Results: Overall 126 out of 160 patients (78%) agreed to complete the electronic questionnaire. A total of 102 patients (81%) completed the item on the DCP. Of these 67 (65%) patients were male and 35 (35%) female. The average age of participants was 62 years (min: 20, max: 85 years). Overall 49% preferred shared responsibility with regards to treatment decision making, 28% preferred to leave the control rather to their physician whereas 22% preferred to be in control about treatment decisions. Higher age of patients was significantly associated with willingness to leave the decision with the physician (p = 0.035). Gender and education were not associated with DCP. Higher levels of distress were significantly associated with delegation of control to physicians (p = 0.038). Quality of life, performance status, nutritional status, intention of treatment and need of psycho-oncological support were not significantly associated with DCP of patients. Conclusion: PROs including patients’ DCP can be elicited as part of routine clinical care. The findings on differing degrees of DCP in patients and possible determinants need to be assessed further on in larger samples, optimally including detailed data on disease and treatment. Physicians at specialist centers may benefit from this information to tailor the decision making process according to needs and preferences of their patients. Disclosure: No conflict of interest disclosed.

Abstracts

P497

Next-generation sequencing to identify driver mutations in cancer of unknown primary Löffler H.1, Kriegsmann M.2, Endris V.2, Pfarr N.2, Weichert W.2, Krämer A.1 Universitätsklinikum Heidelberg, Klinische Kooperationseinheit Molekulare Hämatologie/Onkologie, Deutsches Krebsforschungszentrum und Medizinische Klinik V, Heidelberg, Germany, 2Universitätsklinikum Heidelberg, Pathologisches Institut, Heidelberg, Germany 1

Introduction: Cancer of unknown primary (CUP), which is defined as epithelial malignancy where no primary is detected despite appropriate staging, accounts for 2–3% of all cancers. Histologically, about 90% of CUP tumors are classified as adenocarcinoma or undifferentiated carcinoma. Current management of CUP is focused on identification of the site of origin and, if sensible, administration of site-specific therapy. However, even when using site-of-origin assignment by gene expression profiling, the prognosis of CUP remains poor, with a median overall survival in the range of one year. In an increasing number of other tumor entities, recurrently mutated genes are established as predictive parameters and can be efficiently targeted by specifically designed drugs. Hence, identification of molecular targets might be a promising approach to direct CUP therapy independently of site-of-origin assignment. As yet, scientific evidence regarding the prevalence of oncogenic driver mutations in CUP is scarce, and neither targeted therapies nor predictive molecular markers are established in its treatment. Methods: Tumor DNA was extracted from routine histology samples of patients with either adenocarcinoma or undifferentiated carcinoma meeting the definition of CUP using standardized criteria. Panel sequencing of 50 genes, which had been selected due to their relevance as oncogenic driver genes or druggable molecular targets, was performed using the Ion AmpliSeq™ Cancer Hotspot Panel v2. Results: We here report the preliminary results for the first 33 cases of this study. Tumor-specific mutations were detected in 28 cases (85%). The most frequently mutated genes were TP53 (11 cases, 33%), SMAD4 (4 cases, 12%), HER2/neu (2 cases, 6%), and KRAS (2 cases, 6%). In single cases, mutations were found in APC, ATM, CDH1, CDKN2A, EGFR, ERBB4, FGFR1, JAK3, or MET. In addition, coverage analysis revealed 2 cases of CDKN2A deletions, 2 cases of MET amplifications, and 2 cases of NRAS amplifications. Single cases harbored amplifications of CSF1R, EGFR, or FGFR1. Conclusion: Although mutations of relevant driver genes are present in the vast majority of CUP tumors, the frequency of targetable molecular alterations appears to be low. As in many other tumor entities, the most frequently mutated gene is TP53. Disclosure: No conflict of interest disclosed. P498

Antitumor activity of lanreotide autogel 120 mg in enteropancreatic Neuroendocrine Tumour (NET) patients: the clarinet open-label extension study Pavel M.1, Raderer M.2 Charité, Campus Virchow Klinikum, Berlin, Germany, 2AKH Wien, Wien, Austria

1

Introduction: CLARINET core study data has demonstrated the efficacy and safety/tolerability of lanreotide as an antitumor treatment in patients with metastatic enteropancreatic NETs. Here, we report updated PFS data and safety findings from the CLARINET open-label extension (OLE; NCT00842348). Methods: In the core study, patients with metastatic well/moderately differentiated non-functioning enteropancreatic NETs, Ki-67 <10%, no prior medical therapy in last 6 months were randomized to lanreotide Autogel 120 mg (n = 101) or placebo (n = 103) for 96 weeks or until progressive disease (PD) / death. In the OLE, patients with stable disease (SD) at corestudy end or progressive disease (PD) on core-study placebo entered single-arm OLE on lanreotide. The primary objective was to evaluate longerterm safety of lanreotide; a secondary objective was to investigate further

Oncol Res Treat 2015;38(suppl 5):1–270

151


CONTENTS AUTHOR INDEX

lanreotide efficacy based on PFS (i.e. time from randomization in core study to PD/death) for the core study intent-to-treat population, using survival analysis based on Kaplan-Meier estimates. Results: 88 patients from the core-study (41 on lanreotide, 47 on placebo) entered the OLE. At core-study enrolment, 96% of OLE patients had SD; 38% had pancreatic and 39% midgut primary tumours. For patients continuing on lanreotide, median PFS reached during the OLE was 32.8 months (median PFS in subgroups were: 42.8/29.7 months, midgut/pancreas; 42.8/29.7 months, grade 1/2; 42.8/24.1 months, HTL≤/>25%). For patients who switched from placebo to lanreotide after PD in the core study, median time to further PD with lanreotide in the OLE was 14.0 months. During the OLE, 66% who continued lanreotide vs 81% who switched to lanreotide experienced adverse events (of which, 27% vs 40% were treatment-related); most frequent events were diarrhea. No new safety concerns were identified during the OLE. Conclusions: The OLE study further demonstrates favourable lanreotide safety/tolerability in enteropancreatic NETs. It also provides data on the continued antitumor effects in patients with SD on lanreotide in the core study, and on antitumor effects in patients after PD with placebo. Disclosure: Marianne Pavel: Advisory Role: IPSEN; Financing of Scientific ­Research: IPSEN; Other Financial Relationships: IPSEN. Markus Raderer: Advisory Role: IPSEN; Financing of Scientific Research: IPSEN; Expert Testimony: IPSEN; Other Financial Relationships: IPSEN. P499

Effects of an RGD-peptide in osteoclast maturation and behavior Bianconi D.1, Chillà A.1, Geetha N.1, Dorda A.1, Poettler M.1, Unseld M.1, Sykoutri D.2, Redlich K.2, Zielinski C.C.1, Prager G.W.1 Department of Medicine I, Medical University of Vienna, Vienna, Austria and Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria, 2Division of Rheumatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria 1

Metastatic bone disease is a common burden in many types of cancer and has a severe impact on the quality of life in patients. Hence, specific therapeutic strategies inhibiting tumor induced osteolysis are urgently needed. In this study, we aimed to interfere with integrin adhesion receptors, which are central players of the bone resorption process, including osteoclastogenesis as well as osteoclast/bone matrix interaction. For this purpose, we used cilengitide, a cyclic RGD peptide, which blocks integrin αVβ3 and αVβ5-ligand binding. Our results revealed that cilengitide blocked osteoclast maturation in a dose-dependent manner. In detail, pre-osteoclasts treated with cilengitide exhibited reduced cell spreading, cell migration and cell adhesion on RGD-containing matrix proteins, such as osteopontin and fibrinogen, which are ligands of integrin αVβ3. The activation of the most upstream signal transduction molecules of the integrin receptor-initiated pathway, FAK and c-Src, were consistently blocked by cilengitide. First evidence suggests that cilengitide might interfere with metastatic bone disease in vivo and this study describes a potential underlying mechanism of the inhibitory effect of cilengitide on αV-integrin expressing pre-osteoclasts by blocking integrin ligand binding and interfering with osteoclast maturation and cell behavior. In conclusion, our findings suggest that cilengitide, which interferes with αV-integrins on osteoclasts, may represent a novel therapeutic strategy in the treatment of malignant bone disease. This work was supported by Merck Serono. *A.C. and D.B. contributed equally. Disclosure: Daniela Bianconi: No conflict of interest disclosed. Gerald Prager: Expert Testimony: Das Projekt wurde zum Teil von Merck Serono finanziert.

152

Oncol Res Treat 2015;38(suppl 5):1–270

P500

Treatment of patients with refractory metastatic solid tumors according to a molecular characterization of potential predictive biomarkers Seeber A.1, Gastl G.1, Ensinger C.2, Forcher V.2, Rinderer F.1, Willenbacher E.1, Willenbacher W.1, Eisterer W.1, Pall G.1, Leitner C.1, Spizzo G.3, Amann A.1, Lang A.4, Jaeger T.4, Voss A.5, Zwierzina H.1 Medizinische Universität Innsbruck, Hämatologie und Onkologie, Innsbruck, Austria, 2Medizinische Universität Innsbruck, Pathologie, Innsbruck, Austria, 3 Krankenhaus Meran, Hämatologie und Onkologie, Meran, Italy, 4Krankenhaus Feldkirch, Innere Medizin, Feldkirch, Austria, 5Caris Life Sciences, Basel, Switzerland 1

Introduction: Patients with refractory metastatic cancer have been shown to benefit from molecular profiling of tumor tissue. The “ONCO-T-PROFIL” project was launched in March 2014 at the Department for Haematology and Oncology of Innsbruck Medical University. Within 2 years our project aims to recruit 100 patients with stage IV cancer. Our data presented here is based on an interims-analysis. Methods: Formalin-fixed tumor tissue specimens are submitted for molecular profiling to a certified laboratory in the USA (CARIS Life Intelligence©). Profiling methods used to identify and characterize potential predictive biomarkers include IHC, NGS and CISH/FISH. Druggable tumor targets and corresponding drug candidates were selected based on an evidence-based information system that associates the biomarker status to agents with potential clinical benefit or potential lack of benefit. Clinical benefit was defined as a PFS ratio (=PFS upon the last therapy/PFS upon treatment according to the molecular profile) ≥1.3. Results: Until April 2015, tumors from 50 patients have been molecularly profiled and in 48 (94%) one or more druggable target structures were detectable. So far, 19 (38%) patients have been treated or are currently undergoing treatment according to their molecular tumor profile. To date, 7 of 19 (36%) patients have benefitted by reaching a PFS ratio of ≥1.3. Conclusions: Up to now a subset of our patients showed a clinical benefit from a therapeutic regimen based on molecular typing. Our data demonstrate that a subgroup of patients can benefit from an individualized treatment approach based on molecular profiling. Disclosure: Andreas Seeber: Advisory Role: Beratungstätigkeit für CARIS Life Sciences. Heinz Zwierzina: Advisory Role: Beratungstätigkeit für CARIS Life Sciences. P501

Long term survival after stem cell transplantation Schroeder M.1, Wieschermann U.1, Aul C.1 HELIOS St. Johannes Klinik, Medizinische Klinik 2, Duisburg, Germany

1

Introduction: In 1996 we started among others in 3 pts. with metastasized solid tumors a treatment including stem cell transplantation. High dose chemotherapy (HD-CTX) with autologous stem cell transplantation (auto-SCT) is an established treatment modality for pts. with hematological neoplasms. During the nineties HD-CTX and autologous SCT was also used as adjuvant treatment in pts. with localized breast cancer. Methods: In our department we administered HD-CTX and autologous TX to 3 female pts. with solid tumors, who all exhibited multiple metastases at diagnosis: 1. with breast cancer: 45y, T3 N1 M1 (Lnn) 2. with ovarian cancer: 37y, T3 N1 M1 (pulmo) 3. with SCLC: 34y, T3 N2 M1 All pts. gave written informed consent to this individual treatment approach which was different from the standard of care at this time. Following conventional induction chemotherapy autologous PBSCT were administered. Nextly all 3 pts. underwent a tandem HD-CTX consisting of Carboplatin/Etoposid and autologous TX in 1996. Results: All 3 pts. with metastasized disease achieved a CR. Following this treatment. only one of them also receiving a prophylactic irradiation. At

Abstracts


CONTENTS AUTHOR INDEX

the time of writing all 3 pts. are alive and remain in long term remission without further treatment. Conclusion: Although HD-CTX and autologous SCT is not a standard option for pts. with metastasized gynecological and lung cancer, the course of disease reported were suggests an individual benefit of HD-CTX in our 3 pts. Disclosure: No conflict of interest disclosed. P502

Stem cell markers are overexpressed in tumorspheres cultured from circulating epithelial tumor cells (CETCs) in patients with solid cancers Zimon D.1, Pizon M.1, Maier V.2, Pachmann U.2, Pachmann K.2 SIMFO GmbH, Bayreuth, Germany, 2Labor Dr. Pachmann, Bayreuth, Germany

1

Background: Solid malignancies continuously shed tumor cells which may enter the circulation, spread to other tissue and initiate metastases, but only a small subpopulation among CETCs is capable of metastasis formation. The subpopulation of CETCs capable of forming tumorspheres in vitro and carrying stem cells properties is assumed to be metastatic cells. The aim of this study was to characterize the pattern of expression of the tumorspheres as compared to CETCs. Methods: Blood samples were obtained from patients diagnosed with solid tumors. CETCs were enumerated with maintrac method and subsequently cultivated in stem cell-selective medium. The expression of stem cell marker genes EpCAM, Nanog, Oct4 Sox2, ALDH and CD133 were detected by RT-PCR in single CETCs and tumorspheres. Results: 0.1–10% of CETCs could form non-adherent tumorspheres under stem cell- selective conditions after a period of 14 days. Array qRTPCR analysis revealed that putative stem cell markers, such as Oct4, Sox2, Nanog, EpCAM, ALDH1 and CD133 are overexpressed in relation to house-keeping genes RPL13a and GAPDH in tumor spheres in contrast to the significantly lower expression level of these stem cell markers in individually isolated CETCs. High expression level of pluripotency genes in tumorspheres was associated with aggressive tumor behaviour in terms of tumor progression and type of cancer. Conclusion: Thus, differential expression of stem cell markers in CETCs and tumorspheres may confirm the theory that only a small population of circulating tumor cells possess the stem cell properties and are responsible for metastasis formation. Disclosure: No conflict of interest disclosed.

Results: Up to now, 34 pts were identified (20 male, median age 62 years; 9 pts with solid tumors, 25 with haematological malignancies, 16 pts with active malignant disease at diagnosis of influenza) with community-acquired influenza A (25 pts) or influenza B (9 pts) confirmed by PCR from respiratory samples. Twenty pts presented with upper respiratory symptoms/fever only, 13 suffered from pneumonia at diagnosis and one patient developed pneumonia later on. Six of the pts with pneumonia had active cancer. Unexpectedly, 10 pts also complained of gastrointestinal symptoms. Nine patients had clinically relevant superinfections with invasive fungal infection in 4 pts, bacteraemia in 2 pts and secondary bacterial/ viral pneumonia in 3 pts. Six of the pts with superinfection had active cancer. Thirty-two pts received treatment with oseltamivir, mostly 150 mg/d for a median of 9 days. Four pts required intensive care treatment, two with bacterial and one with fungal superinfection; one of these pts died. One additional patient died before the diagnosis was established and treatment could be administered. Conclusions: The influenza season 2015 was particularly dangerous for cancer pts with a high rate of pneumonia (41%) and -mortality (6%), respectively. Of note, 26% of pts suffered from superinfections leading to a high rate of complications. In conclusion, efforts to diagnose influenza should be reinforced in cancer pts and the high rate of pneumonia and superinfections should influence the management of influenza in cancer pts, particularly when they have active malignant disease. Disclosure: No conflict of interest disclosed.

Posterdiskussion

Versorgungsforschung P504

Analysing the eligibility of chemotherapy agents for dose-banding Reinhardt H.1, Trittler R.2, Wöhrl S.2, Epting T.3, Buck M.4, Kaiser S.1,5, Kaiser S.4, Jonas D.4, Duyster J.1,5, Jung M.6, Hug M.J.2, Engelhardt M.1 University Medical Center Freiburg, Department of Hematology and Oncology, Freiburg, Germany, 2University Medical Center Freiburg, Pharmacy, Freiburg, Germany, 3University Medical Center Freiburg, Department of Clinical Chemistry, Freiburg, Germany, 4University Medical Center Freiburg, Environmental Health Sciences and Hospital Infection Control, Freiburg, Germany, 5Comprehensive Cancer Center, Freiburg, Germany, 6Albert-LudwigsUniversität Freiburg, Institute of Pharmaceutical Sciences, Freiburg, Germany 1

Introduction: Influenza may pose a vital threat to patients with a compromised immune system, but data on epidemiology and effect of treatment in cancer patients (pts) are scarce. In spring 2015 the expected influenza wave was particularly strong, affecting especially the South-East of Germany. We sought to understand the clinical epidemiology and outcome of cancer pts who had documented influenza in 2015. Methods: Data were collected retrospectively on all pts with cancer requiring treatment or a history of cancer who presented with documented influenza infection in two university hospitals (Jena and Magdeburg).

Introduction: In times of increasingly complex requirements for preeminent patient (pt) care accompanied by high pt turnover, organisational-, quality- and financial aspects play a pivotal role. In an interdepartmental, cross functional cooperation, we are investigating the feasibility and benefits of implementing dose-banding (DB) in chemotherapy (CTx) at our medical center. In DB, CTx-doses are clustered into bands of similar dosage levels within a certain range. The mid-point dose of each band represents the respective prescribed dose. This concept elegantly allows the preproduction of frequently used doses for CTx-substances with adequate stability data. Over recent years, DB has become common practice in many UK oncology centers and has shown to be safe and economic, whereas in Germany or other countries, the DB model has not been introduced as yet. Methods and Results: From analysis of local prescribing practice gemcitabine, 5-FU Bolus and carboplatin were identified as most suitable CTx-preparations for DB. For the selected substances physical and chemical stability tests for a maximum storage period of 3 months were performed, results are shown in table 1. Our quantitative analysis showed that after a storage period of 12 weeks ≥95% Gemcitabine was remaining, which is in accordance with international guidelines. In order to simulate the worst case, microbiological stability tests were performed with liquid media instead of CTx: samples were stored for 3 months and incubated at different time points. Microbiological stability could be shown for the complete storage period. In the

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

P503

Influenza in cancer patients – characteristics and outcome of the spring season 2015 from two university hospitals Schalk E.1, Hermann B.2, Meckel K.3, Rachow T.3, Hochhaus A.3, von Lilienfeld-Toal M.3,4,5 Otto-von-Guericke University Magdeburg, Medical Centre, Department of Haematology and Oncology, Magdeburg, Germany, 2Universitätsklinikum Jena, Institut für Medizinische Mikrobiologie, Jena, Germany, 3Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Onkologie, Jena, Germany, 4 Universitätsklinikum Jena, Centre for Sepsis Control and Care (CSCC), Jena, Germany, 5Hans-Knöll-Institut, Nationales Referenzzentrum für invasive Mykosen, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie, Jena, Germany 1

153


CONTENTS AUTHOR INDEX

Tab. 1. zu P504 Physical and chemical stability – results

weeks 0–12

pH-changes

Colour

Particles

Quantitative analysis

Gemcitabine

<0,1

no change in colour

no particles

≥95% after 12 weeks

<0,05

opalescent, no change in colour

no particles

in progress

no change in colour

few floating particles*

in progress

*Announcement of the Federal Institute for Drugs and Medical Devices (BfArM) from 10.07.2012: Recommendation to filter certain Carboplatin-containing drugs with a 0.2 µm filter

5-FU

Carboplatin

<0,2

additional container integrity test the stored infusion bags and syringes were inserted into a highly contaminated broth for 1 hour. After incubation and smear no bacterial growth was found. Conclusions: Storage of cytotoxic preparations over a long period of time (3 months) without contamination is possible, as the results of our microbiological analysis clearly indicate. Even under extreme conditions, when inserted into a bacteria bath, the contents of the preparations remained sterile. Advantages, such as workflow optimisation for pharmacy departments and reduction of patient waiting time, without compromising patient safety, are convincing arguments for dose banding. Disclosure: No conflict of interest disclosed. P505

Measuring communication skills in an oncology ward Wittke G.1, Junghanss C.2 Arbeits-Psychologische Praxis Dr. Gregor Wittke, Berlin, Germany, Universitätsmedizin Rostock, Klinik III, Hämatologie, Onkologie, Palliativmedizin, Rostock, Germany 1 2

Introduction: In the field of oncology the physician-patient interaction may be even more important than in other disciplines. Therefore continuing medical training of communication skills has moved into focus. Whereas medical schools address these issues increasingly during early medical education concepts for clinical oncologists are rare. Here, we report on a pilot project that was launched in the Rostock university hospital to evaluate physicians’ communication skills, implement a quality management approach to ensure high professional skill levels and offer individual coaching for further improvements. Methods: To assess the level of the physicians’ communication skills an observational study was used. The observation criteria were developed based on current research findings. The observations were rated on 39 skills using a newly developed five-point rating scale. The subsequent coaching sessions were based on these results. The present study included 16 physicians of the oncology and palliative care ward, which is a full survey of the physician staff at that point of time (2013). The sample consisted of the head, four senior physicians, two specialist physicians and nine assistant physicians. Each physician was escorted for one day by the evaluator (trained psychologist). Direct feedback was followed by graded results afterwards and an anonymous team report. Results: The Rating scale used to quantify the observations reached an acceptable internal consistency (Cronbach´s Alpha) of 0.7. The result is 25% of the staff met the targeted skill level defined prior to conducting the observation already before the coaching sessions. Some skills showed high levels over all the staff (e.g. listening), some showed a broad spread and others were low over all. That means the observation as well as the rating scale appear to be sensitive to interpersonal differences and deliver reliable scores that can be used to measure changes in the staffs’ communication-skills. A first norm sample was raised. Conclusions: The developed observation criteria and the expert ratings are a valid instrument to assess communications skills in an oncology and palliative care ward. Thus the developed and tested structured approach can be useful to raise methodologically sound measures which provide useful key figures for a quality management which also includes relevant

154

Oncol Res Treat 2015;38(suppl 5):1–270

psycho-social skills. The effect of the coaching sessions will have to be evaluated in a further study. Disclosure: No conflict of interest disclosed. P506

Comparison of data quality in Baden-Württemberg with joined data from all other German epidemiological cancer registries Friedrich S.1, Hermann S.1, Haug U.1, Becker N.1 Deutsches Krebsforschungszentrum, Epidemiologisches Krebsregister Baden-Württemberg, Heidelberg, Germany 1

Background & Introduction: Baden-Württemberg (BW) was the last federal state in Germany to establish a cancer registry and the first to only except electronic reports. After three years run time of state-wide compulsory reporting in BW we aim to compare the quality of the electronically collected data with the data from all other German epidemiological cancer registries, joined at the German Centre for Cancer Registry Data (ZfKD). Materials and methods: We focused on data quality in terms of socio-demographic and clinical information that is available for new cancer cases reported to the cancer registry. We considered cancers diagnosed in the year 2011 and ICD-10 C00-C96 without C44. We assessed the completeness of the following essential variables: gender, birth date (month/year); specific histological codes (not 800–800, 8010); T-, N-, and M-specification (not TX, NX, MX); treatment and generated UICC-stage. We first determined the completeness of information regarding these variables for BW and then compared it with the joined data from all German cancer registries (GCRs), excluding data from BW. Results: The overall number of cancers considered for this analysis was 37.816 for the registry of BW and 415.982 for all other GCRs. Gender as well as date of birth was reported for 100% of the recorded cancers in both datasets. Specific histological information was available for 86.6% and 86.0% of the cancers reported to the registry of BW and all other GCRs, respectively. Details such as T- (64.9% vs. 58.7%), N- (58.7% vs. 47.7%), and M-specification (58.3% vs. 47.0%), were reported less frequently to the registry of BW than to the other GCRs. Due to this, the UICC stage could only be generated for approximately half of the cancers (histology: 800–867, 872–879, including DCO-cases for GCR) recorded in the registry of BW (53,7%) and in the other GCRs (47,0%). At least one information concerning treatment was available for 71,3% of cancers reported to the registry of BW and for 52.9% of cancers reported to the GCRs. Conclusion: There is need to further improve the completeness of clinical data reporting to cancer registries in Germany. Both quality assurance of cancer care and the evaluation of cancer control programs requires detailed information on histological codes, T-, N-, and M-specification, and treatment. Health care relevant research questions can only be addressed if this information is transmitted to the cancer registries as a matter of routine. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

P507

18 months of “mobile oncologic service“ in Lower Bavaria – a new path of ambulant care for tumour patients Kaiser F.1, Jedraßczyk M.2, von Olnhausen A.2, Damnali G.3, Haas M.3, Vehling-Kaiser U.3 Universitätsmedizin, Göttingen, Germany, 2Kassenärztliche Vereinigung Bayern (KVB), München, Germany, 3Onkologisches und Palliativmedizinisches Netzwerk Landshut, Landshut, Germany 1

Introduction: The “mobile oncologic service – MOD“ was established as a model of care for multimorbid, oncologic patients in the rural area. Highly specialized and trained assistants “Qualified person for oral and subcutaneous antitumour-therapy” take care of the supply of oral and subcutaneous antitumor-agents at patients´ homes. Main issues of the project contain transmitting of delegable, medical tasks and improvement of patients´ compliance. Quality of life, safety of therapies and patient satisfaction were considered as indicators. Methods: The sample period of this descriptive study ranged from 08/2013–02/2015. Inclusion criteria were: malignant disease, considerably restricted mobility, high danger of infection and missing options for transport. The number of patients, the frequency of visits, the travel distance for the MOD-assistants and the length of visit per patient were recorded by standardized case report forms. The influence of MOD on quality of life, safety of the therapy and patient satisfaction were registered by a standardized questionnaire and rating scales, ranging from 1–6. Results: Within 18 months, 97 patients were looked after in 791 ambulant visits. Per Quarter an average number of 32 patients were taken care by 2–3 MOD assistants at an average of 4–5 visits at home. The mean distance per visit was 20 km, the mean duration of the visit was 63 minutes (24 minutes of travel, 29 minutes visiting time and 5 minutes of conferring a doctor and documentation, respectively). Overall, 65 patients were interviewed for evaluation: 92% of patients rated with “very good” for the reduced efforts for visiting a doctor´s practice, 58% for the reduction of distress for their relatives and 78,5% for an improvement of their quality of life. 92% of patients assessed the care by MOD as “very important” and 95% were very satisfied with MOD´s work. Conclusions: The additional, ambulant care at home of selected hematologic/oncologic patients by specialized and trained assistants supports a high patient satisfaction. In special, long distances and frequent visits at a doctor´s practice can be avoided for multimorbid patients, substantially improving their quality of life. Compliance and therapy safety can substantially be improved. Two-weekly visits at home – with justifyable effort – seem to be lasting.

pleteness index Clark’s C (Lancet 2002;359:1309) and the modified completeness index Wu’s C* (Ann Thorac Surg 2008;85:1155). Results: A total of 89 patients with CNSL were included (79 primary CNSL). Median age was 67 years (range 23–83). Majority of patients (n = 77) received high-dose methotrexate as first-line treatment. Median follow-up was 5.1 months (range 0.2–117.7) for the standard follow-up (study duration = 14.0 years). At the time of data collection, 59.6% of the data were censored. Clark’s C and Wu’s C* were 45.7% and 60.4%, respectively. For the “pro-active” data inclusion, median follow-up was 7.3 months (range 0.2–170.0) with 28.1% censored data (study duration = 14.3 years). Clark’s C and Wu’s C* were higher with 74.0% and 89.0%, respectively. Of note, median overall survival (OS) using standard follow-up was 33.4 months (95% CI 14.8–52.0) in contrast to the “pro-active” follow-up where median OS was only 9.6 months (95% CI 1.3–17.9) (hazard ratio = 1.56 [95% CI 1.03–2.35]; p = 0.03). Moreover, the 2-year OS was 55.3% and 41.4% for the standard follow-up and the “pro-active” follow-up, respectively. Conclusions: A “pro-active” follow-up with higher completeness may significantly influence overall survival data. The prevailing problem of no follow-up data in clinical trials can lead to bias conclusion. Therefore, quality parameters like censored events and completeness C or C*, respectively, should be stated in publications of clinical trials to improve clinical decision making based on survival data. Disclosure: No conflict of interest disclosed. P509

Interim analysis of the SENSe-study structured evaluation of sustainability of sports after cancer Roggenhofer S.1, Wörtz I.1, Widmann T.2 Waldklinik Dobel, Innere Medizin / Onkologie, Dobel, Germany, 2Asklepiosklinik Triberg, Innere Medizin / Onkologie, Triberg, Germany 1

Introduction: Loss of follow-up is problematic in most clinical studies, since this can severely compromise study´s external validity. Number of patients lost in follow-up is one indicator of data incompleteness. Completeness indices were developed in order to better access data that take into account follow-up years and anticipated deaths. However, little is known how completeness and follow-up can influence overall survival data. Methods: This single-centre, retrospective analysis included all patients with central nervous system lymphoma (CNSL) treated in our institution between 11/2000 and 11/2014. We compared survival data obtained by standard follow-up and data obtained by a “pro-active” follow-up. The latter means an update of patient data that were lost in follow-up by – e.g. – contacting patients’ primary practitioners, family members or nursing homes. To quantify the completeness of follow-up we calculated com-

Introduction: Scientific data exist that among patients who achieved a certain dose of physical activity (18–27 metabolic equivalents per week (MET)), disease reccurency and potentially mortality after cancer can be reduced. We investigate the following questions within the SENSe-study: are all cancer patients able to achieve their medically necessary “dose” of physical activity during an inhouse, 3 week medical rehabilitation process? Can an exercise diary and training plan for at home help to achieve the necessary “dose” of physical activity in the long term. Methods: We started a prospective, randomized, controlled, blinded study in 2014. For the interim analysis in April 2015 n = 182 patients were included. The sample is divided into two groups (A/B), both are interviewed about their previous physical activity by using a standardized questionnaire (German PAQ50+). During rehabilitation patients receive an exercise diary to document their physical activity. In contrast to group B, group A receives an exercise diary/training plan for at home for the next 6 months. Group A and B both are interviewed again about their physical activity after 3 and 6 months by using the above standardized questionnaire. Results: All patients achieved a mean of 23.82 MET/week (SD = 27.15) of physical activity before rehabilitation. During medical rehabilitation, patients achieved a mean of 41.47 MET/ week (SD = 13.51, t = 8.70, p < 0.001), after 3 months a mean of 43.26 MET/week (SD= 27.26, t=-7.37, p < 0.001) and after 6 months a mean of 44.16 MET/week (SD = 32.09, t=4.62, p < 0.001) and thus had a significantly higher score. Interestingly, post-hoc comparisons revealed that 6 months after rehabiltation there was a significant difference between both groups (t = 2.21, p = 0.030). Group A scored more MET per week (M = 54.00, SD = 36.75) than group B (M = 36.72; SD= 26.17). Conclusions: Patients seem to be active before rehabilitation. During inhouse rehabilitation, patients reach prognostic relevant levels of physical exercise and can be guided to keep this level after rehabilitation. Patients receiving additional support by training plan/exercise diary can reach a higher level of physical activity even 6 month after rehabilitation. The aim of any oncological rehabilitation facility should be, to motivate and guide

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. P508

A “pro-active” follow-up with higher completeness can significantly influence survival data Zeremski V.1, Koehler M.1, Fischer T.1, Schalk E.1 Otto-von-Guericke University Magdeburg, Medical Centre, Department of Haematology and Oncology, Magdeburg, Germany 1

155


CONTENTS AUTHOR INDEX

patients to physical activity in the long term in order to reach a positive effect on cancer. This is a very important factor of a sustainable medical rehabilitation. Disclosure: No conflict of interest disclosed. P510

Effects on treatment duration and compliance of patients with metastatic colorectal cancer (mCRC) treated with Aflibercept in combination with FOLFIRI by intensified care with requests for patient-reported-outcomes (PRO) and early interventions to side effects under real-world-conditions in Germany Lipp R.1, Freigang F.1, Feuerbach M.1, Brecht P.1 GermanOncology GmbH, Hamburg, Germany

1

Introduction: In the pivotal trial (Van Cutsem E et al.: JCO 2012,30(28):3499–3506) therapies with Aflibercept+FOLFIRI (A-FOLFIRI) in patients with mCRC were permanent discontinued in up to 26,8% due to adverse events. The aims of this cohorts study were to describe the effects of intensified care on treatment duration and compliance by requesting PROs (QoL,VAS) and early intervening to defined toxicities (diarrhea, hypertension, fatigue, infections) in the real-world-evidence in Germany. Patients with mCRC and A-FOLFIRI were analysed in 2 cohorts: cohort 1 with standardized requests for PROs and early toxicities and cohort 2 without these requests. Results: 62 patients with mCRC from 18 oncological practices throughout Germany (42% women, 58% men, mean age 71 years) were treated with A-FOLFIRI (1°line 3 pat., 2°line 22 pat., 3°line 20 pat., 4°line 9 pat., ≥ 5°line 8 pat.). In cohort 1 (n = 24) patients got requests for QoL (FACT + module for CRC) before start of every cycle and a diary for requests of daily performance (visual analog scale from 0–100) and specific toxicities, in cohort 2 (n = 48) patients were guided during A-FOLFIRI based on oncologist´s choice. During A-FOLFIRI in 85,4% of patients an uptake in QoL scores and in daily VAS was noted compared to baseline for cohort 1. A-FOLFIRI was given over all lines for a mean of 7,3 cycles (14,6 weeks) in cohort 1 and 6,4 cycles (12,8 weeks) in cohort 2. Defined severe toxicities were transmitted to the oncologist in a mean of 2,2 days after first appearance and in 4,8 days, respectively. The rates for permanent discontinuations of A-FOLFIRI due to toxicities were 25,4% in cohort 1 and 34,5% in cohort 2. Dose reductions for A-FOLFIRI were performed for 18,5% in cohort 1 and 21,3% in cohort 2. Conclusions: Requests for QoL/VAS and the intention to early intervention in case of toxicities could be tools to intensify the care and enhance the compliance of patients with mCRC under a treatment with A-FOLFIRI under real-world-conditions. With such intensified care the rates of permanent discontinuations and dose reductions of A-FOLFIRI could be reduced and rates of transmisson of severe side effects to the oncologists and so early anticipations could be improved. An intensified care by requesting PROs and specific toxicities may be a good opportunity for oncological practices to improve the compliance of patients in oncologiucal practices and to reduce permanent discontinuations due to side effects. Disclosure: No conflict of interest disclosed. P511

Evaluation of data mining algorithms for survival prediction Sailer F.1, Bochum S.2, Pobiruchin M.1, Schramm W.1, Martens U.M.2 Hochschule, Heilbronn, Germany, 2SLK-Kliniken Heilbronn GmbH, Tumorzentrum Heilbronn-Franken, Heilbronn, Germany 1

Survival time prediction at the time of diagnosis is of great importance to make decisions about treatment and long-term follow-up care. Cancer survival rates and median survival times are estimated from a large group of cancer patients and mostly based on the TNM-staging system. While these estimates do apply to the population in general, they are not particularly accurate for individual patients. Incorporation of further var-

156

Oncol Res Treat 2015;38(suppl 5):1–270

iables like age and sex – as implemented in most nomograms – improves prediction accuracy. However, since machine learning techniques have the potential to detect hard to discern patterns in complex datasets, data mining algorithms trained on big, national datasets might be the next step towards improving survival time prediction. Methods: We examined the ability of ten different data mining algorithms (e.g. Naïve Bayes, k-nearest Neighbor, Neural Network, Random Forest) to predict the outcome of the dichotomous attribute “5-year-survival – yes / no” based on seven attributes: Sex, ICD-code, number of tumors, UICCstage, histologic type, grading and age at diagnosis. These attributes were selected from a colorectal cancer dataset provided by the Robert Koch Institute (RKI). After pre-processing n = 14,133 cases remained, with half of the cases used as training dataset. A comparison between the algorithms’ accuracy, accuracy of nomogram predictions and physicians’ opinion all based on the same dataset and attributes was performed for 200 randomly selected cases. Results: The average accuracy of the prediction by the participating physicians was 59% (minimum: 54%, maximum: 66%), positive predictive value (PPV) of the physicians’ opinion ranged from 35% to 50%, negative predictive value (NPV) ranged from 66% to 82%. While the achieved prognostic accuracy of nomogram-based prediction and physicians’ opinion were similar to each other, eight out of ten data mining algorithms achieved a higher accuracy than physicians and nomograms, with Random Forest being the most accurate algorithm (accuracy: 79.58%, PPV: 83.99%, NPV: 60.06%). Conclusions: Even though due to the structure of the RKI dataset only a limited number of attributes could be utilized, our data provide significant evidence, that data mining algorithms could improve survival time prediction. In an attempt to further improve prognostic accuracy, we now intend to use de-identified records from a clinical cancer registry that provides considerably more clinicopathological parameters. Disclosure: No conflict of interest disclosed. P512

Data requirements in centralised marketing authorisation procedures versus benefit assessment of cancer drugs Schubert A.1, Tebinka-Olbrich A.1, Zentner A.1, Haas A.1 GKV-Spitzenverband, Arznei- und Heilmittel, Berlin, Germany

1

Introduction: Several mechanisms such as orphan drug designation, conditional marketing authorisation and marketing authorisation under exceptional circumstances exist, that aim at accelerating the assessment process in order to fasten patient access and stipulate research and development. This trades off the necessity for evidence versus faster access. However, data requirements are different for the assessment of additional benefit by the Federal Joint Committee (FJC) than for marketing authorisation, which may impact on the verdict of the FJC regarding the question of the existence, probability and extent of additional benefit. This project tries to clarify the impact of different pathways and data requirements of marketing authorisation on the process of benefit assessment. Methods: Empirical data presented by manufacturers of cancer drugs for marketing authorisation, summarised in the European Public Assessment Report, in benefit dossiers and the benefit assessment report by IQWiG or the FJC are summarised in terms of the study design, measures applied, their validity and statistical uncertainty of results. The findings are contrasted with the way the FJC considers the data as adequate and patient relevant. Reasons given by the FJC in the written statements for incorporating or neglecting evidence related to the data are presented. Results: Data provided for new active oncological substances authorised under alternative procedures often pose significant challenges for the quantification of the probability and extent of the additional benefit by the FJC. This relates to surrogate parameters, the questionnaires used for the assessment of quality of life, and endpoints such as progression-free survival and often leads to the verdict of a “non-quantifiable” additional benefit or a lower probability.

Abstracts


CONTENTS AUTHOR INDEX

Conclusion: At present, there is a tension between the principles of confirmatory trials, the tools to enhance patient access and the principles of evidence-based medicine underlying health technology assessment or early benefit assessment. EMA is currently developing the concept of “adaptive pathways” as part of its efforts to improve timely access for patients to new medicines. The consequences of this new approach for early benefit assessment will need to be discussed. Disclosure: No conflict of interest disclosed. P513

Patient preferences for overall survival and chemotherapy side effects in lung and colon cancer Schmidt K.1, Damm K.1, von der Schulenburg J.-M.1, Studiengruppe an der Medizinischen Hochschule Hannover: Prof. Dr. med. T. Welte und die Klinik für Pneumologie; Prof. Dr. med. M. P. Manns und die Klinik für Gastroenterologie, Hepatologie und Endokrinologie Center for Health Economics Research Hannover (CHERH), Leibniz Universität Hannover, Hannover, Germany 1

Introduction: There is a growing interest in improving patient relevant outcomes during cancer treatment – besides the clinical parameters. Because severe side effects may occur the patients should be included in the decision making process regarding their treatment. Patient preferences can provide further insights into the importance of attributes and side effects that influence health related quality of life (HRQOL). Furthermore, we gain information about which side effects patients would accept for prolonged survival. In Germany, the leading causes of cancer deaths are lung cancer (LC) and colon cancer (CRC). The aim of this study is to identify the treatment preferences of LC and CRC patients. Methods: The attributes for the Discrete Choice Experiment (DCE) were identified in guided qualitative interviews (n = 38). LC and CRC patients aged 18 years and older who received at least one cycle of chemotherapy are eligible for inclusion. In the DCE, two hypothetical treatments of different attribute levels are compared. Sociodemographic, HRQOL, and disease specific data are collected. Descriptive statistics and multivariate regression methods (conditional logit and latent class models) will be employed. Results: The preliminary sample comprises of 57 patients (32 LC, 25 CRC) with a median age of 64 years. For treatment of LC and CRC, the most important attributes are: survival, guidance through therapy, appearance, physical capacity as well as food intake/digestion. The conditional logit model for choice of therapy showed a significant association with survival (Odds Ratio = 8.31, p < 0.001), physical capacity (OR = 0.46, p < 0.001), food intake and digestion (OR = 0.45, p < 0.01) as well as appearance (OR = 0.81, p = 0.05). Additional analyses will be conducted after completion of patient recruitment. Conclusion: Overall survival seems to be the most important attribute for treatment. These findings are consistent with current state of research. Additionally, this study provides first indications about the LC and CRC patients´ willingness to trade quality for quantity of life. Further results will be updated during the next months until completion of the recruitment of 250 LC and 250 CRC patients. Also subgroup analysis of LC and CRC patients will be conducted. A Limitation of this study is the focus on chemotherapy patients. However, first recommendations for patient oriented improvement of therapy can be drawn. Disclosure: No conflict of interest disclosed.

Abstracts

P514

The “Taking Care Project”: A special program for complex cancer patients in rural areas Weiglein T.1, Vehling-Kaiser U.2, Tischer J.1, Hiddemann W.1, Kaiser F.2 Medízinische Klinik III, Universitätsklinikum der LMU, München Großhadern, Hämatologie und Onkologie, München, Germany, 2Onkologisches und Palliativmedizinisches Netzwerk Landshut, Landshut, Germany 1

The “Taking Care Project” (TCP) is a collaboration of the Oncologic-Palliative-Care-Network of Landshut and the University Clinic of Oncology and Hematology of the Ludwig Maximilians University in Munich, Germany. The TCP was implemented to improve the medical care for cancer patients in remote rural Areas via different approaches. Firstly, giving inbound patients from Landshut to the university hospital a specially trained “care taker” as a chaperone who is an employee of the clinic and will take care of the patient for the whole duration of the stay. This is achieved by regular independent visits of the “care taker”, including problem solving, confidence building and psychological help. Standardized questionnaires are regularly answered by the patients to detect problems early on and to generate data for the scientific part of the TCP. The care takers also coordinate admissions and discharges, apply for access to tumor boards and complex therapies etc. Secondly, all patients are admitted to a specific ward as well as to a specific doctor in order to minimize loss of information and to streamline the overall process. In addition specialist consultations by senior university physicians – e.g. on bone marrow transplantations, thoracic surgery, orphan disease and pain management – are made possible by establishing the consultations on-site in the rural practice. Another integral part of the TCP is the ongoing yearly rotation of physicians in training from the University Hospital to the practice in Landshut, enhancing the knowledge transfer between the University Hospital and the oncological practice both ways. This possibility of a job rotation serves as a strong incentive for young doctors to work in rural areas and gain hands-on experience otherwise not possible within a university hospital. When all the individual factors are successfully put together, they permit to improve the quality of life and standard of care for the affected patients and their families. This project also optimizes the work flow for all involved staff and has been proven economically feasible. Preliminary results of standardized questionnaires, Show following improvements: “better medical standard”, “less time lost on the road”, “less anxiety”, “more trust” and “less distress”. The long term goal is to implement the setup of the TCP to other regions and institutions. The TCP is currently financially supported by the Bavarian State Office for health- and food safety. Disclosure: No conflict of interest disclosed. P515

Total cost per year associated with “Best Supportive Care“ of patients with unresectable advanced non-small-cell lung cancer (NSCLC) under “real-world-conditions“ in relation to results of international studies Feuerbach M.1, Freigang F.1, Lipp R.1, Zentren der GermanOncology Gruppe GermanOncology GmbH, Hamburg, Germany

1

Introduction: Only a few international studies give valid information about cost of BSC of patients with unresectable advanced or metastatic NSCLC. The aim of this non interventional study was to compare results of these international studies with data collected under “real-ife-conditions“ in 26 oncological practices in Germany. Methods: Data from the 26 oncological practices in Germany contain information about the entire course of treatment including e.g. medication of primary disease, radiotherapy, number of outpatient specialist visits, hospitalisations, number of diagnostic and laboratory procedures. In addition four different studies from USA, Scotland, Great Britain and the Netherlands with information about cost of BSC were examined. To compare these studies with data from the 26 oncological practices in Germany,

Oncol Res Treat 2015;38(suppl 5):1–270

157


CONTENTS AUTHOR INDEX

the results were transfered into the German reimbursement systems for health care services. The calculation for the annual costs of treatment is based on the methods of the IQWIG. Results: 509 patients with NSCLC from 26 oncological practices throughout Germany (38% women, 62% men, mean age 69 years) were included in the statistical and cost´s analyses. Patients had to be in unresectable stage IIIB or stage IV and under current anticancer treatment at enrollment (independent of lines of treatment). The calculations included direct costs for all oncological therapies, supportive and concomitant medication, involvement of other medical disciplines, hospitalisations (other than administration of chemotherapy), transfusions and other more. The total cost of illness for these patients were in median 10,098.00 € per patient per year. In contrast, the median total cost of BSC in the international studies were 29,621.48 € (range from 17,365.16 € to 50,882.80 €). Conclusions: Due to lack of information from studies about the cost of BSC treatments for NSCLC in Germany, data collected under “real-world-conditions“ gets more relevant for BSC analysis. This analysis shows clinical and cost data of 509 patients with NSCLC treated in 26 oncological practices throughout Germany. The results of this analysis illustrate a significant difference in the annual costs from data under german real-world-conditions in relation to the median cost from four international studies. A main reason for the lower total cost compared to the referenced studies could be the higher share of less expensive outpatient services in the german health system.

A significant correlation between household income and financial burden was found (r = –0.498, p < 0.001), while the correlation with personal income was not significant (r = –0.188, p = 0.112). Only 29% of all participants reported a satisfying level of information concerning public financial support in case of unemployment and cancer. Conclusion: Diagnosis and treatment of cancer regularly result in additional costs for patients which for some pose a serious obstacle in addition to coping with the illness itself. Offers of information on public financial support in case of unemployment and illness have to be extended.

Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

P516

Tab. 1.

N

Income [M(SD)]

Houshold income [M(SD)]

Experienced burden [M(SD)]

Full time work

22

2077(781)

3362(1727)

2.56(2.76)

Part-time work

11

753(373)

2943(1396)

2.56(2.64)

Unable to work

20

1573(1185)

2852(1565)

4.77(3.24)

Unemployed

5

780(305)

760(482)

5.94(4.74)

Househusband/ -wife

4

67(115)

1967(473)

1.3(1.26)

Pensioner

14

1190(724)

2003(1161)

3.56(2.89)

P517

Financial impact of cancer on industrial workers – first results of a systematic survey

Mobilizing stem cells with plerixafor: Impact on a health care providers budget

Peters E.1, Reuss-Borst M.1

Berger K.1, Jensen I.S.2, Gallagher M.E.3, Ostermann H.1

Rehabilitation Clinic for Rheumatology and Oncology, Bad Kissingen, Germany

1

Introduction: As a result of current structural changes in working life and health care systems an increasing number of patients confronted with a long lasting (chronic) disease describe their financial situation as a main psychological stress factor. The aim of this study was to systematically investigate the effects of a cancer disease on the financial situation of a large population of industral workers and craftsmen. Methods: Between 12/2014 and 02/2015 all N = 180 oncological patients were given a self developed questionnaire concerning their financial situation via internal mail. To grant anonymity in the evaluation, patients were asked to put the filled questionnaires back in an internal post box as well. The questionnaire consists of five parts labeled A) Current (work and household) Situation, B) Situation before Cancer, C) Direct financial impact of Cancer, D) Means of financial adaptation, E) Information on financial support. 86 questionnaires (48%) were handed back in. Results: Participants were M = 55 years of age (SD = 10.2). Breast cancer was the most common diagnosis (49%, n = 42). 43% of participants were working at assessment. Average personal net income was 1344€ net, household income was 2663€. Participants rated their financial burden at M = 3.5 on a scale from 1 to 10. Table 1 summarizes incomes and financial burden for participants with different sociomedical status.

158

Oncol Res Treat 2015;38(suppl 5):1–270

Klinikum der Universität München, Medizinische Klinik und Poliklinik III, München, Germany, 2ICON Plc, Health Economics, Blue Bell, United States, 3 Sanofi, Global Health Economics & Outcomes Research, Global Market Access Center of Excellence, Cambridge, United States 1

Introduction: Autologous stem cell transplantation following high dose therapy has become a routine procedure for myeloma and many lymphoma patients. Autologous harvest of stem cells from the peripheral blood is achieved following a mobilization procedure. Mobilization treatment has consisted of chemotherapy and the administration of G-CSF. In 2009, plerixafor, a CXCR4 antagonist, was approved for use in combination with G-CSF to enhance this procedure, allowing more patients to mobilize sufficient quantities of stem cells. As plerixafor introduces an additional cost to hospital budgets, we analyzed the annual budget impact of adding plerixafor for stem cell mobilization in multiple myeloma (MM) and lymphoma patients compared to G-CSF ± chemo. Methods: A budget impact model based on literature data and KOL input was adapted to a German health care provider perspective. Costs included the first and re-mobilization. Drug costs were taken from official German list prices (excl VAT) and hospital operating costs were applied from a tertiary German hospital. Clinical inputs were obtained from clinical experiences at a tertiary hospital. The base case analysis assumes the pre-plerixafor era mobilization use and the scenario case includes the availability of plerixafor in combination with G-CSF and chemotherapy. Results: Model calculations simulate 50 patients with MM and 15 lymphoma patients. For patients with MM and lymphoma, total plerixafor use increased from 0–18% and 0–26% respectively, leading to a 5.6% increase in successful mobilizations for MM and a 14.2% increase for lymphoma. Plerixafor acquisition costs are offset by lower laboratory costs, pre-apheresis costs, peri-apheresis costs, storage costs and hospital stays. The annual budget increase due to plerixafor introduction is estimated to be 9.6% (€ 1451/patient) for MM and 13.9% (€ 2656/patient) for lymphoma. Conclusions: This budget impact analysis from a German health care provider perspective suggests that use of plerixafor in patients with MM and lymphoma may result in better clinical outcomes (more successful first mobilizations with fewer remobilizations) and is associated with a min-

Abstracts


CONTENTS AUTHOR INDEX

imal increase of the provider’s budget. These budget calculations do not take into account local hospital reimbursement agreements with plerixafor. Additionally, for a comprehensive health economic assessment, treatment impact on patient quality of life should also be considered. Disclosure: Karin Berger: Expert Testimony: Research grant from ICON. Helmut Ostermann: Expert Testimony: Research grant from ICON. P518

Cost of illness in patients with pancreatic tumor treated under “real-world-conditions“ in 26 oncological practices in Germany Feuerbach M.1, Freigang F.1, Lipp R.1, Zentren der GermanOncology Gruppe GermanOncology GmbH, Hamburg, Germany

1

Introduction: The cost of illness in patients with pancreatic tumor treated completely under outpatient´s setting in oncological practices are often difficult to calculate as mostly not all data of every treatment line, concomitant diseases and other cost relevant factors are available. The aim of this non interventional study was to calculate the total cost of illness and to analyse this distribution of costs to the relevant cost drivers. Methods: Data included in this study contain information about the entire course of treatment including the medication of primary disease, radiotherapy, number of outpatient specialist visits, length of hospitalisation and other more. The calculation for the annual costs of treatment is based on the methods of the IQWIG. Results: 614 patients with pancreatic tumor from 26 oncological practices throughout Germany (47% women, 53% men, mean age 70 years) were included in the statistical and cost´s analysis. The first given therapy was dated in 2002, the last in April 2015. The calculations included direct costs for all oncological therapies, supportive and concomitant medication, involvement of other medical disiplines, hospitalisations, transfusions and other more. The total cost of illness for this group of patients were in median 97,339.67 € per patient per year. This total costs are dispensed on 77,267.19 € (=79.4%) for antitumor medication (cytostatics and monoclonal antibodies), 1,377.31 € (=1.4%) for oncological services, 932.87 € (=1.0%) for concomitant medications, 11,620.93 € (=11.9%) for hospitalisation, 103.04 € (=0.1%) for radiotherapy, 2,424.55 € (=2.5%) for supportive medications, and 299,54 € (=0.3%) for other services like laboratory or rehabilitation. Conclusions: This analysis shows data of 614 patients with pancreatic tumor treated under real-world- and purely out-patient´s conditions in oncological practices throughout Germany. The annual cost of illness for a patient with pancreatic tumor has been calculated at in median 97,339.67 € over all lines of treatment whereas more than 79% of these costs have been caused by antitumor medication including cytostatics and monoclonal antibodies. In contrast, costs for oncological services like elucidations of treatments, controls and observations during treatment, management of side effects etc. were calculated at 1,377.31 € (=1.4%) which proved the presently enhancing shift in the costs from oncological care towards pharmaceutical drugs. Disclosure: No conflict of interest disclosed. P519

COMunication with oncological patients – essential for therapeutic adhereNCE – COMPETENCE Musch R.1, Lück A.2, Rubanov O.3 Krebsheilkunde Lichtenberg, Berlin, Germany, 2MVZ für Onkologie und Urologie, Rostock, Germany, 3Hämatologische und Onkologische Praxis Rubanov, Hameln, Germany 1

Introduction: One goal of the German national cancer plan ist goal 12b, “strengthening of the patient´s competence” by offering information, support and advice as well as optimized conditions and development of stimuli for professional support of patient´s competence. In this context professional skills of specialized (oncological) nurses is of particular im-

Abstracts

portance. To find out the conditions regarding patient´s competence in specialized oncological centers in Germany we performed a nationwide survey. Methods: 30 specialized oncological centers (hospital and office based) in Germany participated in this project. The online-survey covered general information about the centers and the patient population, information transfer to patients, role of specialized oncological nurses, and – as a model for patient´s competence – a detailed questionaire about supportive therapy with (daily applied) G-CSF. The survey was performed from October 2014 for 9 months. Results: The participating centers were mainly office based hemato-oncological or gynecological specialists. The patients´ average age was 65 years, average weight was 76 kg and body surface was 1,9 m². 65% were retired and their diseases were dominantly solid tumors (75%), among them lung cancer (26%), breast cancer (13%) and prostate cancer (11%) were the most common. In 50% of the centers at least 20% of all patients receiving G-CSF receive daily applied G-CSF – mainly because of effectivity and suitable dosage. For information of patients the specialized oncological nurses play a central role in more than 50% of the centers. About 70% of the patients show average interest in additional information considering their disease or (selfapplication of) therapies. Considering the daily G-CSF therapy with lenograstim, about 75% of patients use self application and only about 20% of patients prefer application by nursing care or specialized nurses in the centers. In more than 90% of chemotherapy cycles 5 or less daily applications of lenograstim were regarded as sufficient. Conclusion: The survey revealed an important role of specialized oncological nurses in patient information to strengthen patient´s competence. Well informed patients develop a good competence to support the therapeutic efforts by e.g. self application of daily G-CSF which is effective and cost-efficient. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium

Lunge / Pleura personalisierte Therapieansätze V521

NSCLC: Rare molecular subtypes Gautschi O.1 Kantonsspital Luzern, Tumorzentrum, Luzern, Switzerland

1

Introduction: Molecular targeted therapy is a new standard of care for patients with advanced non-small cell lung cancer (NSCLC) with activating EGFR mutations or ALK translocations. Further druggable oncogenic drivers are known, but their incidence is lower, which has implications for clinical research and drug approval. Methods and Results: This lecture will cover the clinical value of ROS1 and RET translocations, and HER2 and BRAF mutations, in patients with advanced NSCLC. Key topics will include molecular discovery and epidemiology, clinical diagnostics, preliminary experience with targeted therapy in individual patients, results from first prospective phase I-II trials, and retrospective cohort studies including EUROS1 (Mazieres JCO 2015), EUHER2 (Mazieres 2013) and EURAF (Gautschi ELCC 2015). Conclusions: The testing of advanced lung adenocarcinomas for rare molecular alterations is clinically relevant, because individual patients can derive substantial clinical benefit from targeted therapy. Pathologists and clinicians are faced with new challenges when dealing with the interpretation of test results and having to allocate patients to targeted therapies. Academic collaboration at the national and international level is necessary to make further progress in this exciting field. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

159


CONTENTS AUTHOR INDEX

Fortbildung

How I treat PV and ET in 2015

vera are defined by the WHO criteria. Those who do not fulfil these criteria require further investigation depending on the clinical scenario and initial results. The erythropoietin level provides some guidance as to the direction in which to proceed and the order and extent of investigation necessary in an individual patient. It should thus be possible to make an accurate diagnosis in the majority of patients.

Lengfelder E.1

Disclosure: No conflict of interest disclosed.

MPN Chronische Myeloproliferative Erkrankungen V526

Universitätsmedizin Mannheim, Hämatologie/ Onkologie, Mannheim, Germany 1

Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPN), respectively characterized by erythrocytosis and thrombocytosis. Other disease features typically include leukocytosis, splenomegaly, pruritus and in more advanced disease fatigue. The natural course is characterized by thrombohemorrhagic complications and a propensity to transform into myelofibrosis and acute leukemia. The description of the JAK2 V617F mutation has not only improved the understanding of the pathophysiology of the MPN, but has also provided a target for specific therapy. Approximately 98% of PV patients have a mutation in the JAK2 gene. In ET, mutations in the JAK2 gene, the calreticulin gene and the MPL gene occur in approximately 60%, 25% and 3% of patients, respectively. Median survival is over 15 years in PV and approximately 20 years in ET, which is inferior to the normal population. In both diseases, the most frequent and potentially dangerous complications are arterial or venous thromboses. The thrombotic risk increases with older age and the presence of thrombosis. Both factors therefore represent the basis of risk stratification of therapy (low risk: no risk factor, high risk: one of the two or both risk factors). The main goal of therapy of PV and ET is to prevent the thromboembolic complications. For low risk PV patients treatment with low dose acetylsalicylic acid (ASS) and phlebotomy (hematocrit target < 45%) is recommended. Low dose ASS is also indicated in low risk ET patients with microvascular disturbances. High risk PV and ET patients should be treated with hydroxyurea (HU) or interferon alpha (younger individuals). Second line approaches are ruxolitinib (PV) and for elderly patients busulfan. The administration of experimental therapy within clinical trials can be considered in the case of treatment failures. Ruxolitinib represents the first JAK2 inhibitor, which has recently been approved for the treatment of patients with HU resistant or intolerant PV on the basis of the results of an international randomized trial (RESPONSE). In this trial, patients who had an inadequate response to or had unacceptable side effects from HU, ruxolitinib was superior to the best available therapy in controlling the hematocrit and spleen volume. Furthermore, ruxolitinib improved symptoms associated with PV impressively. Disclosure: Eva Lengfelder: Advisory Role: Advisory Board Novartis Pharma. GmbH; Other Financial Relationships: Reisekostenerstattungen Novartis Pharma GmbH. V527

Differential diagnosis of erythrocytosis

Fortbildung

Konfrontation oder Kooperation: Interaktion von Onkologen und Organonkologen V533

A model of cooperation between outpatient private practices of urologists and oncologists Schmitz S.1, Grund C.1, Jülicher H.1, Finke F.1, Klier J.1, Nazari S.1, Maus R.1, Severin K.1, Steinmetz H.T.1 AFO fachübergreifende ambulante Onkologie Köln, Köln, Germany

1

In 2014 a joint cooperation was launched by six urologists and three hematologists and oncologists with the conviction that bringing together both expertises would grant a higher quality of medical treatment. This innovative cooperation between oncologists and oncological oriented urologists is based on the analysis which reveals a significant change of political surrounding conditions and requirements of modern medicine over the last few years. Therefore the founders aim is to organize an interdisciplinary network of outpatient private practices in order to provide an ideal offer for their patients. We describe the legal structure, the organization, the infrastructure and professional interaction of all parties. Another goal is to benefit from economic synergies, to optimize process, develop a new range of services offered and consolidate them on a professional-political basis. Altogether the expectations of this organization are to strengthen the position of contractual physicians in perspective of the increasing competition in the range of ambulant oncology. Also this cooperation could be an example of further interdisciplinary oncological cooperation. Disclosure: No conflict of interest disclosed.

Freier Vortrag

AML experimentell II V537

Significance of the IL-3 receptor beta chain (IL-3Rβc) for FLT3-ITD dependent oncogeneic signaling in AML Rummelt C.1, Gorantla S.P.1, Kläsener K.2, Keye P.1, Rao V.1, Reth M.2, Duyster J.1, von Bubnoff N.1

Johannes Wesling Klinikum, Minden, Germany

Uniklinik Freiburg, Innere Medizin, Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 2Max-Planck-Institute for Immunobiology and Epigenetics, Freiburg, Germany

The differential diagnosis of erythrocytosis is a clinical relevant problem in internal medicine. Erythrocytosis results when there is an increased red cell mass and thus an increased hemoglobin and/or haematocrit. The causes can be divided into primary intrinsic defects of the erythroid progenitor cell and secondary defects, where factors external to the erythroid compartment are responsible. Both can then be further divided into congenital and acquired categories. Congenital causes include mutations of the erythropoietin receptor and defects of the oxygen-sensing pathway including VHL, PHD2 and HIF2A mutations. There remain an unexplained group idiopathic erythrocytosis. Investigation commencing with thorough history taking and examination and then investigation depending on initial features is required. Clear simple criteria for polycythaemia

Introduction: Activating FLT3 mutations are found in 30% of AML patients. Internal tandem duplication (ITD) mutations are most common, and are associated with poor prognosis. Clinical studies with FLT3 kinase inhibitors show limited activity and treatment resistance arises rapidly. In our quest to identify mechanisms of treatment-resistance in FLT3-ITD positive AML, we identified a direct interaction between FLT3-ITD and IL-3Rβc. We therefore sought to further characterize this interaction and determine its’ role in FLT3-ITD oncogenic signaling. Methods: Drug resistant sublines were generated by exposure of stably transfected BA/F3 FLT3-ITD cells to FLT3 kinase inhibitors (TKIs). These sublines were selected for further analysis by PCR, Western-Blot (WB) and IP. Interaction studies in human cells were performed in MOLM13,

1

Griesshammer M.1 1

160

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

MV4–11 cells and primary AML patient samples via WB, IP and PLA. Knockdown-experiments were performed using shRNA against IL-3Rβc in BA/F3 cells. Results: In TKI resistant FLT3-ITD positive cell-lines without a secondary FLT3 resistance mutation, we observed phosphorylation of IL-3Rβc in 30% (5/16). This phosphorylation was mediated by an activating JAK1 mutation that bypassed FLT3-ITD dependent IL-3Rβc phosphorylation. Of note, we observed a “physiologic” phosphorylation of IL-3Rβc by FLT3-ITD via direct interaction of the two proteins in TKI sensitive cells, demonstrating that IL-3Rβc participates in FLT3-ITD dependent oncogeneic signaling. In order to query the relevance of our findings in a human system, we could confirm the interaction using human constructs in γ2A cells and in human AML cell-lines MOLM13 and MV4–11. Proximity ligation assay (PLA) experiments revealed localization of FTL3-ITD and IL-3Rβc in close proximity, suggesting a direct interaction of both molecules in MOLM13 and MV4–11 cells, as well as in primary FLT3ITD positive AML patient samples. Finally, we performed IL-3Rβc knock down experiments in FLT3-ITD expressing BA/F3 cells, demonstrating that knock-down cells were more susceptible to TKI inhibition. Conclusion: IL-3Rβc directly interacts with and is phosphorylated by FLT3-ITD in murine and human cells. TKI resistant FLT3-ITD positive cell-lines use JAK1/2 dependent IL-3Rβc phosphorylation to bypass FLT3-ITD as escape mechanism. Knocking down IL-3Rβc results in increased susceptibility to TKI treatment. These findings point toward the relevance of IL-3Rβc as a possible target in FLT3-ITD positive AML. Disclosure: Christoph Rummelt: No conflict of interest disclosed. Nikolas von Bubnoff: Financing of Scientific Research: Novartis, BMS. V538

Incidence and clinical relevance of ASXL2 mutations in adult AML with t(8;21)(q22;q22): A study of the German-Austrian AML Study Group (AMLSG) Jahn N.1, Bullinger L.1, Weber D.1, Corbacioglu A.1, Gaidzik V.1, Agrawal M.1, Schlenk R.1, Thol F.2, Heuser M.2, Krauter J.3, Göhring G.4, Kündgen A.5, Fiedler W.6, Lübbert M.7, Ganser A.2, Döhner H.1, Döhner K.8, Paschka P.1 Klinik für Innere Medizin III, Universitätsklinikum, Ulm, Germany, 2Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Germany, 3Medizinische Klinik III, Klinikum, Braunschweig, Germany, 4Institut für Humangenetik Medizinische Hochschule Hannover, Hannover, Germany, 5Klinik für Hämatologie, Onkologie und Klinische Immunologie, Universitätsklinikum, Düsseldorf, Germany, 6II. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany, 7Klinik für Innere Medizin I, Universitätsklinikum, Freiburg, Germany, 8Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany 1

Background: ASXL2 (Additional Sex comb-like 2) gene on chromosome 2p23.3 encodes a protein thought to act as a chromatin modifier. ASXL2 mutations were recently reported with a high frequency (~23%) in a cohort of 110 pediatric or adult AML cases with t(8;21)(q22;q22) (Micol et al., Blood 2014). In our study we assessed the frequency of ASXL2 mutations and their prognostic relevance in the context of other clinical factors and genetic markers in a large cohort of adults with t(8;21) AML. Methods: ASXL2 mutation status was analyzed in pre-treatment samples from 204 adults (median age: 49 years (yrs), range: 18–82) with t(8;21) AML. Most patients (pts) were intensively treated within AMLSG trials (n = 155); all other pts received intensive chemotherapy according to standard regimens. ASXL2 regions in exons 11 and 12 spanning the main mutational clusters were analyzed by DNA-based PCR-amplification and subsequent direct DNA-sequencing. Results: 34 ASXL2 mutations were detected in 33 (16.2%) of 204 pts; all mutations resulted in protein truncation (frame-shifts, n = 32; nonsense, n = 2). There was no significant difference between ASXL2 mutated (ASXL2mut) and ASXL2 wildtype (ASXL2wt) pts in terms of age, sex, and clinical baseline characteristics including WBC, haemoglobin, platelets, LDH, circulating or bone marrow blasts. With respect to secondary chromosome aberrations ASXL2mut were frequently associated with del(9q) (P=.006), whereas they never co-occurred with trisomy 8. Moreover, there

Abstracts

was no significant association of ASXL2mut with other gene mutations including KIT, FLT3 (ITD and TKD), and NRAS. The complete remission rate after double induction was similar for pts with ASXL2mut (88%) and those with ASXL2wt (92%); the same was true when comparing patients with ASXL1 or ASXL2 (ASXL1/2mut) mutations as one group (88%) and pts with ASXL1/2wt. Finally, neither ASXL2mut nor ASXL1/2 mut did impact on event-free-survival, cumulative incidence of relapse, relapse-free survival and overall survival. Conclusions: In this large cohort of t(8;21)-positive AML we identified ASXL2mut in 16.2% of the pts representing beside KITmut one of the most frequently mutated genes in this AML subtype. The high frequency of ASXL2mut and the exclusivity to this core-binding factor AML subtype (Micol et al., 2014) suggest a peculiar involvement in the leukemogenesis of AML with t(8;21) providing a basis for further studies. Disclosure: No conflict of interest disclosed. V539

Functional roles of the transcription factor CDX2 in human acute myeloid leukemia Paczulla A.1, Konantz M.1, Quintanilla-Martinez L.2, Kanz L.3, Lengerke C.1,4 Department of Biomedicine, University Hospital, Basel, Switzerland, Department of Pathology, University of Tuebingen, Tuebingen, Germany, 3 Department of Hematology & Oncology, University of Tuebingen Medical Center II, Tuebingen, Germany, 4Clinic for Hematology, University Hospital, Basel, Switzerland 1 2

Introduction: The caudal-type homeobox (CDX) gene family has been mainly studied during early development where CDX genes were shown to regulate embryonic hematopoiesis via downstream HOX genes and interactions with the WNT signaling pathway. Healthy bone marrow (BM) derived hematopoietic cells express low levels of CDX1 and CDX4 but lack CDX2 expression. However, CDX2 expression is found in >80% of human acute myeloid (AML) and lymphoid leukemia (ALL) and its induction in murine BM cells results in myeloid leukemia. Here, we explore the role of CDX2 in human healthy hematopoietic and leukemic cells. Methods: CDX2 expression was modulated in human leukemic cell lines and BM CD34+ cells via treatment with lentiviruses containing CDX2 shRNA or overexpression constructs. Clones displaying a particularly strong knockdown were individually grown from single transduced cells. CDX2-modified and control cells were subjected to growth, colony forming (CFU), cell cycle, adhesion on stromal cells, flow cytometry and qRTPCR assays and analyzed in vivo upon subcutaneous and intravenous xenotransplantation in NOD/SCID/IL2Rγnull (NSG) mice. DKK-1 protein levels were measured in the supernatant via ELISA. Results: CDX2 knockdown strongly reduced AML cell clonogenic capacity in CFU assays while not grossly influencing cell proliferation. Importantly, in vivo leukemia initiation was strongly suppressed in NSG mice transplanted with CDX2 knockdown versus control AML cells. Moreover, adhesion assays using MS5 stromal cells revealed lower adhesion capacity of CDX2 knockdown cells compared to control cells. Molecularly, the expression of HOX and Wnt-pathway associated genes were influenced following CDX2 modulation. Surprisingly, CDX2 induction enhanced the expression of the WNT-inhibitory molecule DKK-1 while CDX2 suppression showed opposite effects reducing also secreted DKK-1 levels. Supplementation of DKK-1 rescued the clonogenicity of CDX2 knockdown leukemic cells in CFU-assays while, upon application in vivo, suppressing healthy hematopoietic stem/progenitor cells. Conclusions: Our data suggest that CDX2 regulates in vivo leukemogenesis by inducing clonogenic properties and adhesive capacity to the BM in human AML cells. CDX2-positive AML cells may use DKK-1 secretion to fine-tune their Wnt-signaling activity to an optimal dosage that enables leukemia initiation but inhibits healthy HSCs thus conferring AML cells a competitive advantage for bone marrow niche occupation. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

161


CONTENTS AUTHOR INDEX

V540

Attenuation of Apoptosis-stimulating protein of p53–2 (ASPP2) facilitates a monosomal karyotype with loss of tp53 and acquisition of additional structural abnormalities in gamma-irradiated hematopoietic cells – mimicking early leukemogenesis of therapy-associated AML Kampa-Schittenhelm K.1, Mau-Holzmann U.2, Kanz L.1, Schittenhelm M.M.1 Universitätsklinikum, Tübingen, Germany, 2Universitätsklinikum Tübingen, Institut für Medizinische Genetik, Tübingen, Germany 1

Inactivation of tp53 by chromosome/gene loss or loss-of function mutation is frequently observed in therapy-associated acute myeloid leukemia. Tp53 mutations associate with complex and monosomal karyotypes and provide the strongest predictive risk factor in this subgroup. ASPP2 is a haploinsufficient tumor suppressor, which is attenuated in acute leukemia via methylation by unknown mechanisms. We now provide evidence that attenuation of ASPP2 may act as an early leukemia-initiating event – facilitating loss of its binding partner tp53 and acquisition of structural aberrations. mRNA and protein screens of ASPP2 expression levels confirmed preferential low levels in complex karyotype AML, arguing for a role of dysfunctional ASPP2 in leukemogenesis. To evaluate for malignant transformation of ASPP2-knock out (ko) cells, long term cell cultures with stably retrovirally silenced, IL3-dependant murine Ba/F3 cells (+/– stress inforcement using gamma irradiation, 2 × 5Gy) were employed. Notably, only ASPP2(ko) cell strains were successfully weaned for IL3 – an indicator of autoactivated cellular proliferation and highly suggestive for acquisition of oncogenic mutations. In contrast, empty vector controls did not survive the weaning process. Cytogenetic analysis of the chromosome set of parental (+IL3) vs. irradiated empty vector controls (+IL3) and ASPP2(ko) cell strains (–IL3) were performed. Interestingly, gamma irradiation did not significantly alter findings for the empty vector controls compared to the parental cells – but demonstrated a consistent chromosomal loss of chr8, der8 and der19 – and, most intriguingly, loss of chromosome 11 (which locates tp53 in mice), down to a monosomal karyotype. In addition, several marker chromosomes (6–8 in ASPP2i vs. 0–2 in parental cells) were identified. We are now in the process of verifying our findings in knockout native bone marrow donor blasts. Notably, our findings not only mimick the pathogenesis of therapy-associated AML – but also of hypodiploid acute lymphoblastic leukemia, which is as well associated with tp53 loss-of-function and predicted for a dismal outcome. Conclusions: Our findings provide rigorous evidence that loss of ASPP2 is a driver mechanism to fuel malignant transformation. Thus our data will shed new light onto relevant questions in basic and clinical oncogenesis of leukemia. Consecutively we will study the reasons for attenuated ASPP2 expression and aim to find means to restore its function. Disclosure: No conflict of interest disclosed. V541

microRNA (miR)-9 directly downregulates the oncogenic transcription factor ets related gene (ERG) & high expression associates with improved outcome in patients (pts) with acute myeloid leukemia (AML) Grimm J.1, Bill M.1, Weidner H.1, Knyrim M.1, Schmalbrock L.1, Jentzsch M.1, Schubert K.1, Wurm A.1, Gerloff D.1, Cross M.1, Lange T.1,2, Behre G.1, Niederwieser D.1, Schwind S.1 Universität Leipzig, Hämatologie und Internistische Onkologie, Leipzig, Germany, 2Asklepios Klinik Weißenfels, Hämatologie und Internistische Onkologie, Weißenfels, Germany 1

Introduction: High ERG expression may be driven by down-regulation of ERG targeting miRs & associates with worse outcome in AML. Methods: 3 miR-9 binding sites (BS) were identified in the ERG 3′-untranslated region (UTR) & analyzed by luciferase assays. We measured

162

Oncol Res Treat 2015;38(suppl 5):1–270

ERG & miR-9 levels in 9 AML cell lines by qRT-PCR. Bone marrow (BM) pri-miR-9 expression in 131 AML pts & 6 healthy individuals was examined. For outcome analyses the highest quartile of diagnostic pri-miR-9 defined high expressers. 2 AML cell lines with low miR-9 & high ERG mRNA levels (KG1a, Kasumi-1) were transfected with a miR-9 expression or empty control vector or treated with Paclitaxel (PTX) -a potential miR9 inducer. In K562 cells (low ERG & high miR-9 expression) knock-down was achieved by anti-miR-9 transfection. We analyzed mRNA & protein level by qRT-PCR & western blot, respectively & performed proliferation & flow cytometry based apoptosis assays. Findings were validated in primary AML blasts (n = 2 pts). Results: Following miR-9 overexpression in 293T cells 1 of the miR-9 BS was confirmed & showed down-regulation of luciferase activity to 52.0% (P=.02), that was rescued by BS mutation. We observed an inverse correlation of ERG & miR-9 levels in AML cell lines (r = –0.9). pri-miR-9 expression was significantly downregulated in BM of AML pts vs healthy individual BM (P < 0.01). miR-9 overexpression in KG1a decreased ERG mRNA & protein level (64.9% & 20.3% after 6h) & reduced proliferation to 53.9% after 4d (P=.01). 10nM PTX upregulated miR-9 to 520% in KG1a cells & decreased ERG mRNA & protein level (51.4% & 7.2%) after 12h. Knock-down of miR-9 in K562 increased ERG mRNA & protein level after 12h (452% & 321%) & proliferation (after 4d to 146%, P = 0.03). miR9 overexpression sensitized KG1a cells to cytarabine (ara-C) treatment, leading to a 19% higher apoptosis rate after 1µM ara-C addition after 48h (P = 0.01). Down-regulation of ERG by elevating miR-9 levels & the associated effects on proliferation & apoptosis were confirmed in Kasumi-1 & primary AML blasts. Finally, high pri-miR-9 expressing AML pts had lower cumulative incidence of relapse (P = 0.04; 26.20% vs 43.90% at 3y) & longer overall survival (P = 0.03; 62.90% vs 38.80% at 3y). Conclusion: Anti-leukemic miR-9 directly regulates ERG expression & higher expression associates with improved survival in AML. Since PTX elevates miR-9 levels a combination with standard chemotherapy may improve pts’ outcome, especially in ERG-driven AML. Disclosure: No conflict of interest disclosed. V542

Quantitative detection of DNMT3A R882H mutation in acute myeloid leukemia Blau O.1, Berenstein R.1, Suckert N.1, Baldus C.1, Pezzutto A.1, Dörken B.1, Blau I.W.1 Charité Universitätsmedizin Berlin, Department of Hematology, Oncology and Tumourimmunology, Berlin, Germany 1

Background: DNMT3A mutations represent one of the most frequent gene alterations detectable in acute myeloid leukemia (AML) with normal karyotype. Although various recurrent somatic mutations of DNMT3A have been described, the most common mutation is located at R882 in the methyltransferase domain of the gene. Because of their prognostic significance and high stability during disease evolution, DNMT3 mutations might represent highly informative biomarkers for prognosis and outcome of disease. Methods: Using new established allele-specific PCR with a Blocking reagent (ASB-PCR assay) for the quantitative detection of DNMT3A R882H mutation, we analyzed 357 follow-up samples from 24 AML patients after therapy and allogeneic stem cell transplantation (alloSCT). Thirteen patients included in the follow-up analysis harbored a NPM1 mutation at diagnosis. This enabled to compare of DNMT3A stability during both CR and complete molecular remission (molCR) using the well-established marker for detection of MRD. In addition, we analyzed FLT3, IDH1, and IDH2 mutations in diagnostic and follow up samples. Results: New developed an ASB-PCR assay for quantitative analysis of R882H DNMT3A mutation displayed a high efficiency and sensitivity up to 10–3. This assay displayed a complete concordance with sequencing and endonuclease restriction analysis. We have found persistence of DNMT3A R882H mutations in complete remission (CR) after standard cytoreduction therapy that could be indicating presence of DNMT3A mutation in

Abstracts


CONTENTS AUTHOR INDEX

early pre-leukemic stem cells that resist chemotherapy. The loss of correlation between NPM1 and DNMT3A in CR could be associated with evolution of pre-leukemic and leukemic clones. After alloSCT in patients in CR with complete donor chimerism we have no found DNMT3A R882H. This data suggests the removal of leukemic stem cells after alloSCT and indicates the importance of alloSCT for high risk AML patients. In relapse of leukemia, all samples showed an increasing of both NPM1 and DNMT3A mutated alleles. DNMT3A mutated alleles increased in relapse after alloSCT and after cytoreduction therapy. This suggests at least in part the presence of NPM1 and DNMT3A mutations in the same cell clone. Conclusion: Quantitative detection of DNMT3A R882H mutations at different time points of AML disease could provide additional information about the role of mutations in development and progression of AML. Disclosure: No conflict of interest disclosed.

Freier Vortrag Stammzellen II V543

Absence of secreted frizzled-related protein 2 (Sfrp2) from the niche leads to overactivation of hematopoietic stem cells under conditions of stress Ruf F.1, Istvanffy R.1, Schreck C.1, Grziwok S.1, Pagel C.1, Peschel C.1, Oostendorp R.1 III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität, München, Germany 1

Members of the Sfrp family are known to bind Wnt family members and inhibit canonical, catenin-mediated Wnt signaling. We found that Sfrp2 was overexpressed in stromal cells which maintain hematopoietic stem cells (HSCs) in culture. Study of Sfrp2–/– mice did not reveal major abnormalities of the hematopoietic system . Nevertheless, lineage-negative Kit+ Sca-1+ (LSK) cells showed a strongly increased number of colony-forming cells in cultures on Sfrp2–/– stromal cells. This was accompanied by increased regeneration of LSK cells with higher levels of Ki67 expression, as well as increased DNA damage response (gH2AX) and epigenetic activation. Yet, total repopulating activity of these cultures was diminished, suggesting exhaustion of HSCs. Since culture is a model of stress, we studied in vivo stress, as induced by 5-FU treatment. Similar to the cultures, 5-FU-treated Sfrp2–/– mice showed increased number of LSks in the bone marrow, 6 days after start of treatment. Furthermore, these LSK cells incorporated more BrdU, expressed more Ki67, and also showed activation of canonical Wnt signaling. Under regenerative stress, transplanted WT HSCs showed increased regeneration of the donor LSk compartment in Sfrp2–/– recipient mice. HSC activation in 1° Sfrp2–/– recipients was accompanied by canonical Wnt signaling (Dvl2, catenin, cyclin D1) as well as non-canonical Nfatc1 activation and translocation. In secondary recipients, the activated HSCs did not perform well, leading to strongly decreased canonical Wnt signaling in 3° recipients with exhaustion of HSC repopulating activity. These experiments show that Sfrp2 expression in the niche is required under stress conditions to limit DNA damage and canonical Wnt-mediated HSC activation. Disclosure: No conflict of interest disclosed.

Abstracts

V544

Connective tissue growth factor (CTGF) is an inducible stroma-derived regulator of normal hematopoietic stem cell regeneration and cell cycle progression Istvanffy R.1, Vilne B.1,2, Ruf F.1, Pagel C.1, Schreck C.1, Henkel L.3, Grziwok S.1, Prazeres da Costa O.3, Götze K.1, Schiemann M.3, Peschel C.1, Mewes H.W.2, Oostendorp R.1 III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, München, Germany, 2Technische Universität München, Institute of Bioinformatics and Systems Biology, Freising, Germany, 3Technische Universität München, Institute of Medical Microbiology, Immunology and Hygiene, München, Germany 1

In culture, HSCs can be preserved with UG26–1B6 stromal cells, or their conditioned medium. Hematopoietic stem cell (HSC) are regulated by their ‘niche’, which limits activation of HSCs, to ensure their maintenance and self-renewal. Gene expression changes of UG26–1B6 stromal cells in co-cultures with Lineage- Sca-1+ Kit+ cells (LSKs) identified connective tissue growth factor (Ctgf) to be upregulated in stromal cells in response to LSK cells. Culture of HSCs on stromal cells with decreased Ctgf (shCtgf) expression iimpaired their engraftment and long-term quality of co-cultured HSCs in vivo. To dissect the underlying mechanisms, we found that CD34- CD48- CD150+ LSK (CD34- SLAM) stromal co-cultures with diminished CTGF increase in G0. In addition, single CD34- SLAM culture in conditioned medium of shCtgf stroma show delayed time to first cell division, which can be rescued by addition of recombinant CTGF. To understand this observation, we assembled a CTGF signaling network model, based on known surface receptors of CTGF expressed by LSK cells (TGFBR1, ITGAV, LRP6), which we then experimentally validated. This showed that CD34-, but not CD34+, SLAM cells express the TGFBR1 receptor, which actvates Smad2/3-dependent signaling, strongly increases CDKN1B expression, and downregulates Cyclin D1. The increase in G0 was further associated with increased DNA damage (gH2AX) and other markers of cellular senescence. Our data shows inducible stromal CTGF upregulation in response to co-culture with LSK cells, which is required to prevent senescence and properly activate cell cycle progression and the maintenance long-term repopulating HSCs in culture. Disclosure: No conflict of interest disclosed. V545

Influence of Di(2-ethylhexyl)phthalate on migration rate and differentiation of human hematopoietic stem and progenitor Cells (CD34+) Caddedu R.-P.1, Manz P.2, Wilk M.1, Fritz B.2, Fischer J.C.3, Haas R.1, Wenzel F.2 Universitätsklinik Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany, 2Hochschule Furtwangen, Medical and Life Sciences, Villingen-Schwenningen, Germany, 3Universitätsklinik Düsseldorf, Institut für Transplantationsdiagnostik und Zelltherapeutika, Düsseldorf, Germany 1

Introduction: Phthalates are a group of synthetic plasticizers that are ubiquitous environmental pollutants with toxic and endocrine disrupting characteristics. DEHP is the most commonly used plasticizer in the world and is still applied to stem cell transfusion bags used for storage of hematopoietic stem and progenitor cells (CD34+ HSPC), which are transferred during stem cell transplantation. Here we examined the effect of DEHP on vitality of CD34+ HSPC as well as stem cell specific properties like migration and differentiation capacity – both important for successful stem cell transplantations. Material and methods: CD34+ HSPC were incubated for 24 h and 72 h with DEHP concentrations ranging from 1 µg/ml to 250 µg/ml. DEHP was diluted in DMSO. Migration rate was analyzed along an SDF1α gradient using Transwell migration inserts. Differentiation of CD34+ HSPC was investigated after two weeks in methylcellulose with colony stimulating factors. Apoptosis rate was measured via Annexin V and 7-AAD staining.

Oncol Res Treat 2015;38(suppl 5):1–270

163


CONTENTS AUTHOR INDEX

Results: 24 h of incubation with 10 µg/ml DEHP led to a significant (p < 0.01) decrease in migration rate of CD34+ HSPC (70.70% ± 7.53%) with a minimum migration rate of 48.33% ± 6.72% in relation to control after incubation with 100 µg/ml DEHP for 72 h. Incubation with the highest tested DEHP concentrations (50 and 100 µg/ml) significantly (p < 0.05) altered colony formation rate and cell type distribution. Apoptosis rate of CD34+ HSPC significantly (p < 0.05) increased after incubation with concentrations of 10 µg/ml DEHP for 24 h (1.46 ± 0.19) with a maximum apoptosis rate of 2.71 ± 0.66 after 24 h incubation with the highest DEHP concentration (250 µg/ml) in relation to control. Conclusion: As shown, DEHP takes impact on migration rate, apoptosis rate, and differentiation of CD34+ HSPC. As these are functions with an important role in stem cell transplantations, the usage of DEHP-free stem cell transfusion bags should be considered. Disclosure: No conflict of interest disclosed. V546

Quality assurance system significantly improves performance of autologous peripheral blood stem cell collection Lisenko K.1, Hundemer M.1, Schmitt A.1, Puthenparambil J.1, Pavel P.2, Hillengass J.1, Witzens-Harig M.1, Goldschmidt H.1,3, Ho A.D.1, Wuchter P.1 Universitätsklinikum Heidelberg, Abteilung Innere Medizin V, Heidelberg, Germany, 2Institut für Klinische Transfusion und Zelltherapie (IKTZ), Heidelberg, Germany, 3Universität Heidelberg, Nationales Centrum für Tumorerkrankungen (NCT), Heidelberg, Germany 1

Introduction: Collection of peripheral blood stem cells (PBSC) by leukapheresis (LP) for autologous transplantation represents a well-established process for many years. However, as a new generation of LP machines has been launched in 2014, comprehensive measures of benchmarking and quality control need to be defined in order to ensure consistent collection performance. Methods: This retrospective study evaluated PBSC collection data of patients with symptomatic multiple myeloma (MM) in first-line therapy (n = 216) or refractory/relapsed Non-Hodgkin lymphoma (NHL) (n = 86) who underwent LP in 2013 and 2014 at our institution. In 2014, we implemented new standard operation procedures (SOPs) for LP including a previously published formula for predicting daily PBSC collection based on CD34+ cell count in the peripheral blood (Rosenbaum ER et al., Cytotherapy 2012). As benchmark, we calculated a performance ratio: collected / predicted CD34+ cells. Approximately half the patients in 2014 underwent LP with the new Terumo BCT Spectra Optia, while the COBE Spectra was used in all other cases. Results: There was no significant difference in collection performance between the Spectra Optia machine and the COBE Spectra. However, in 2014 stem cell collections for MM patients had a significantly higher performance ratio than in 2013 with both machine types (p < 0.006), most probably resulting from the newly implemented SOPs and benchmarking system. For NHL patients the performance ratio in 2013 and 2014 was not significantly different, which might be due to the heterogeneity of this patient group. Conclusions: Introduction of a comprehensive quality management system based on a predictive formula allowed specific adjustment of LP parameters with two types of LP machines. In the group of patients with MM, these measures resulted in a significantly higher collection performance.

Freier Vortrag

Multiples Myelom experimentell V547

The SCFFBXO3 ubiquitin ligase prevents p53-dependent cell death upon DNA damage and promotes the development of multiple myeloma Richter C.1, Engel K.1, Gloeckner J.2, Langer C.3, Peschel C.1, Bassermann F.1 Klinikum rechts der Isar (Technische Universität München), München, Germany, 2NZNE, Tübingen, Germany, 3Universitätsklinikum, Ulm, Germany 1

Introduction: Multiple myeloma cells are characterized by high sensitivity to proteasome inhibitors, indicating an important role of the ubiquitin proteasome system in the pathophysiology of this disease. Genomic instability is a cornerstone of myelomagenesis, underscoring the central role of the DNA damage response machinery. Loss of the tumor suppressor p53 reduces cell death in response to DNA damage and deletion of the p53 chromosomal region specifies a high risk group of myeloma patients. Cellular functions of WT p53, on the other hand, are tightly regulated by post-translational mechanisms, among others by the DBC1 (Deleted in Breast Cancer 1) protein. DBC1 regulates the p53 pathway by direct stabilization of p53 and by inhibition of SIRT1, a p53 deacetylase. Methods: Proteome-wide mass spectrometry was performed to identify potential substrates of the E3 ubiquitin ligase SCFFBXO3. Immunoprecipitation experiments and immunofluorescence microscopy confirmed binding and colocalization of Fbxo3 and binding partners. FBXO3-mediated ubiquitylation was analyzed under denaturing conditions. Biological effects of FBXO3 were assessed by RNAi-mediated knockdown and overexpression. DNA damage was induced by Doxorubicin or γ-irradiation. Experiments were carried out in different multiple myeloma cell lines, HEK 293T, U2OS and Cos7 cells. Expression of FBXO3 in patient samples was determined by qPCR. Results: We identified the ubiquitin ligase SCFFBXO3 as a promising candidate featuring genetic amplification in MM. Specific analysis of FBXO3 gene expression in 129 multiple myeloma patient samples revealed significant overexpression of FBXO3 (29%). Overexpression was found to be mutually exclusive with loss of the p53 tumor suppressor (del 17p13) (p = 0.034). A functional role for FBXO3 in the p53 pathway and in myelomagenesis is further supported by the identification of DBC1 as a potential substrate. Overexpression of FBXO3 increases ubiquitylation of DBC1 upon induction of DNA double strand breaks, whereas knockdown reduces it. Upon prolonged treatment with doxorubicin FBXO3 is upregulated and relocalizes from the cytoplasm to the nucleus to colocalize with DBC1. Accordingly, knockdown of FBXO3 promotes and FBXO3 overexpression prevents DNA damage induced apoptosis. Conclusion: We identified FBXO3 as a potential oncogene in multiple myeloma. Amplification of FBXO3 appears to be a mechanism to inactivate the p53-mediated DNA damage response. Disclosure: Carmen Richter: No conflict of interest disclosed. Florian Bassermann: Expert Testimony: Celgene.

Disclosure: Katharina Lisenko: No conflict of interest disclosed. Patrick Wuchter: Advisory Role: Consultancy, honoraria and membership on Advisory Boards of Sanofi-Aventis.

164

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V548

Final results for the 1703 phase 1b/2 study of Elotuzumab (Elo) in combination with Lenalidomide (Len) and dexamethasone (dex) in patients (pts) with Relapsed/ Refractory Multiple Myeloma (RRMM) Raab M.S.1, Richardson P.G.2,3, Jagannath S.3,4, Moreau P.5, Jakubowiak A.J.3,6, Facon T.7, Vij R.3,8, White D.9, Reece D.E.10, Benboubker L.11, Zonder J.3,12, Tsao L.C.13, Anderson K.C.2,3, Bleickardt E.14, Singhal A.K.13, Lonial S.3,15 Universitaetsklinikum, Heidelberg, Germany, 2Dana-Farber Cancer Institute, Boston, United States, 3Multiple Myeloma Research Consortium, Norwalk, United States, 4Mount Sinai Medical Center, New York, United States, 5University Hospital, Nantes, France, 6University of Chicago, Chicago, United States, 7Hopital Claude Huriez, Lille, France, 8Washington University School of Medicine, St. Louis, United States, 9Queen Elizabeth II Health Sciences Centre, Halifax, Canada, 10Princess Margaret Hospital, Toronto, Canada, 11CHU Tours-Hopital Bretonneau, Tours, France, 12Karmanos Cancer Institute, Detroit, United States, 13 AbbVie Biotherapeutics Inc. (ABR), Redwood City, United States, 14BristolMyers Squibb, Wallingford, United States, 15Winship Cancer Institute, Emory University School of Medicine, Atlanta, United States 1

Introduction: Elo, a humanized IgG1 mAb targeting Signalling Lymphocytic Activation Molecule F7 (SLAMF7) kills myeloma cells with minimal effects on normal tissues. In Ph1 of this open-label dose-escalation study, Elo (5–20mg/kg) + Len/dex ORR in pts with RRMM has previously been reported. [1]. Here we report final results. Methods: Pts with RRMM and ≥1 prior therapy in Ph1 or 1–3 prior therapies (no prior Len) in Ph2 were enrolled (NCT00742560). Ph2 cohort was randomized to 10 or 20mg/kg Elo. Treatment was administered in 28day cycles (Table) until disease progression/unacceptable toxicity. All pts in Ph2 received a premedication regimen to mitigate infusion reactions (IRs). Primary endpoint: ORR (≥partial response) according to IMWG criteria; secondary endpoints included PFS and safety. [1]. Tab. 1. Treatment schedule

Study drug

Dose

Treatment days

Infusion rate

Cycles 1–2 Cycle 3+

Elo

10 mg/kg or 20 mg/kg (IV)

Days 1, 8, 15, 22

Cycles 1–4 up to 2 mL/min; Cycles 5+ up to 5 mL/min (if no IR at 2 mL/min)

Len

25 mg (oral)

Days 1–21

N/A

Dex

28 mg oral + 8 mg (IV) or 40 mg (oral)

On Elo dosing days or weekly

N/A

Days 1, 15

Results: In Ph1, 28 pts were treated (5 mg/kg, n = 3; 10 mg/kg, n = 3; 20 mg/kg, n = 22); median PFS was 33 mo. In Ph2, 73 pts (median age 63 y) were treated (10 mg/kg, n = 36; 20 mg/kg, n = 37). Median no. treatment cycles: 17 (range 1–51). As of 16 Jan 2014, 13 pts in Ph2 (10 mg/kg, n = 6; 20 mg/kg, n = 7) were still on treatment; 60 pts had discontinued, mainly due to disease progression (57%). ORR was 84% (10 mg/kg, 92%; 20 mg/ kg, 76%): a stringent complete response/complete response in 14% of pts, 43% very good partial response, 27% partial response. Median PFS: 29 mo (10 mg/kg, 32 mo; 20 mg/kg, 25 mo). Most common treatment-emergent AEs: diarrhea (66%), muscle spasms (62%), fatigue (56%), and constipation (51%), similar to Ph1. [1] 58% of pts experienced serious AEs, most commonly pneumonia (12%). Of 3412 infusions given, 1127 (33%) were at 5mL/min. Overall rate of IRs was 11%; 7 (10%) pts had IRs at a flow rate ≤2 mL/min, 1 pt (2%) had an IR at >2mL/min. No new AEs were observed at >2 mL/min vs ≤2 mL/min. At the time of analysis 5 deaths were reported across Ph1/2. Conclusions: In this Ph1b/2 study, Elo, an immunostimulatory Ab with a novel mechanism of action, demonstrated clinically meaningful effica-

Abstracts

cy (ORR and PFS) in combination with Len/dex in pts with RRMM. Elo given at rates ≤5 mL/min was well tolerated with low IRs. Ph3 trials are ongoing. Reference: 1 Lonial S, et al.: J Clin Oncol. 2012;30:1953–1959. Disclosure: Marc Raab: Expert Testimony: Celgene, Novartis, Bristol-Myers Squibb, and Amgen; Other Financial Relationships: Consultancy fees from Celgene, Novartis, Bristol-Myers Squibb, and Amgen. Sagar Lonial: Expert Testimony: Millennium, Celgene, Novartis, Bristol-Myers Squibb, and Onyx; Other Financial Relationships: Consultancy fees from Millennium, Celgene, Novartis, Bristol-Myers Squibb, and Onyx. V549

Junctional adhesion molecule-C (JAM-C) mediates adhesion of myeloma cells to bone microenvironment and is involved in cell-adhesion mediated drug resistance Kraus S.1,2, Sancho A.3, Dotterweich J.2, Zeck S.2, Meissner-Weigl J.2, Ebert R.2, Jakob F.2 Universitätsklinikum Würzburg; II. Medizinische Klinik, Würzburg, Germany, Universität Würzburg, Orthopädisches Zentrum für Muskuloskelettale Forschung, Würzburg, Germany, 3Universität Würzburg, Institut für Funktionswerkstoffe der Medizin und der Zahnheilkunde, Würzburg, Germany 1 2

Introduction: Multiple myeloma (MM) is a malignant disorder of post-germinal center B cells and characterized by the clonal proliferation of transformed plasma cells. Complex interactions between the bone marrow (BM) microenvironment and MM cells are crucial for the pathogenesis of myeloma bone disease and provide a protective mechanism favoring survival, proliferation, and migration of MM cells. Importantly, adhesion molecule-mediated interactions are implicated in the development of resistance to cytotoxic drugs. Results: To identify relevant components of BM/MM interactions, we mimicked the BM niche in vitro by co-culturing osteogenic precursor cells (OPC) with the myeloma cell line INA-6. Gene expression profiling identified Junctional adhesion molecules-B (JAM-B) and -C (JAM-C) to be potentially involved in BM/MM crosstalk. qPCR and FACS analyses verified the induction of JAM-B expression in OPC after direct contact with MM cells. We used a blocking antibody against JAM-C to investigate the OPC/MM interaction in a fluorescence-based cell adhesion assay in more detail. Following pre-incubation with anti JAM-C, the strong adhesive interaction was significantly reduced by 37.78% (±18.97%). Remarkably, the application of the antibody after 48h of co-culture was sufficient to disrupt the strong adhesion, indicating that this “active“ mechanism could be exploited to target established cell-cell interactions. Furthermore, we performed functional in vitro assays with regard to proliferation and apoptosis to reveal the putative roles of JAM-B/JAM-C in MM pathophysiology. CellTiter-Glo® 3/7 Luminescent Cell Viability Assay showed a decreased cell viability of INA-6 cells by blocking JAM-C, indicating an active signature remodeling. We next evaluated the effect of JAM-C inhibition on caspase activation in vitro. Whereas the pre-incubation with anti-JAM-C did not affect apoptosis rate, anti-JAM-C in combination with bortezomib induced apoptosis in MM cells in a synergistic manner. These results demonstrate that the disruption of adhesive JAM-B/-C interactions between BM and MM cells enhances the drug sensitivity of MM cells. Discussion: Here, we identified the JAM-B/JAM-C interaction as a relevant component of BM/MM crosstalk. Blocking this interaction in combination with established anti-myeloma therapeutics represents a promising future approach to mobilize niche-protected tumor cells and enhance their sensitivity and vulnerability to cytotoxic therapy. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

165


CONTENTS AUTHOR INDEX

V550

V551

Mapping the impact of proteasome inhibitor therapy on the antigenic landscape of multiple myeloma: Identifying robust targets for T cell immunotherapy

A novel 3D high-throughput coculture platform for ex vivo drug screening in multiple myeloma

Kowalewski D.J.1, Walz S.2, Weisel K.2, Backert L.1,3, Schuster H.1, Kohlbacher O.3, Kanz L.2, Salih H.R.2,4, Rammensee H.-G.1,5, Stevanovic S.1,5, Stickel J.S.2 Interfaculty Institute of Cell Biology, Department of Immunology, Tübingen, Germany, 2University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany, 3University of Tübingen, Department of Computer Science, Tübingen, Germany, 4German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Clinical Collaboration Unit Translational Immunology, Heidelberg, Germany, 5DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Tübingen, Germany 1

Recent studies underscore that multiple myeloma is an immunogenic disease and suggest that it might be effectively treated by T cell based immunotherapy via immunomodulation. Unspecific immune checkpoint inhibition might be synergistically complemented by therapeutic vaccination, which may help induce and guide specific anti-cancer T cell responses. We have recently conducted a study which directly characterized the antigenic landscape of myeloma by mass spectrometric analysis of naturally presented HLA ligands and identified a panel of T cell epitopes characterized by exquisite myeloma-association. As novel immunotherapeutic interventions will have to be investigated as 2nd line treatments, it is of great importance to thoroughly characterize and take into account the effects of previous therapies on the antigenic landscape of target cells. Here we present a longitudinal investigation of the HLA-presented immunopeptidome of myeloma cells under treatment with the 2nd generation proteasome inhibitor carfilzomib. We characterize and quantify the plasticity of the antigenic landscape and identify targets characterized by robust presentation. We quantified HLA surface expression on 4 human myeloma cell lines (MM.1s, U266, RPMI8226 & JJN3) at t0, t24h and t48h after incubation with carfilzomib. Strikingly, we detected elevated levels of HLA class I post treatment for 3/4 cell lines, with absolute molecule counts ranging from 50,000–400,000 molecules/cell. Out of the >5,000 different HLA ligands we could identify on MM.1s and U266 by mass spectrometry, we were able to detect and longitudinally quantify 38 and 54 myeloma-associated epitopes described in previous studies. Importantly, the vast majority of these antigens (88.2%) showed robust presentation on myeloma cells under therapy. Overall, around 12% of HLA ligands showed significant changes in abundance after carfilzomib treatment. Strikingly, we observed a significant reduction in aromatic residues at the C-terminal anchor position, which we interpret as a direct reflection of the mechanism of action of carfilzomib. Our study provides direct insights into the plasticity of T cell antigen presentation on myeloma cells in a model of proteasome inhibitor therapy. We were able to characterize general changes in the immunopeptidome composition and identify robustly presented myeloma-associated epitopes. Our findings may help guide future clinical trials for 2nd line T cell immunotherapy of multiple myeloma.

Waldschmidt J.M.1, Wider D.1, Thomsen A.R.2, Aldrian C.3, Kortüm K.M.4, Follo M.1, Schüler J.5, Duyster J.1, Wäsch R.1, Engelhardt M.1 Freiburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 2Freiburg University Medical Center, Department of Radiation Oncology, Freiburg, Germany, 3Freiburg University Medical Center, Department of Radiation Oncology, Freiburg im Breisgau, Germany, 4Mayo Clinic, Department of Hematology-Oncology, Scottsdale, United States, 5Oncotest GmbH, Department In Vivo Tumorbiology, Freiburg, Germany 1

Introduction: Recently approved new compounds have considerably expanded our treatment options in Multiple Myeloma (MM). However, the development of novel drugs remains inefficient, displayed by a substantial drop out rate of ~400 preclinical single agents tested within the last 50 years (Kortüm, CLML 2014). To better predict clinical success we established and specifically adapted an innovative 3D co-culture model for ex vivo compound screening in MM. Methods: We compared previous 2D and a novel 3D co-culture model (agarose matrix interlayer, 100 microwells/interlayer, 1.5mm in depth, permeable for oxygen+cytokines, but not for BMSCs (Fig.1. A+B) utilizing U266, RPMI-8226, OPM-2 and primary BM patient (pt) cells, w and w/o HS-5 vs. M210B4 stroma support. Analyses covered Trypan Blue, Annexin/PI, MTT, FACS, cell cycle analyses and H2B-mCherry/cytochrome c-GFP assays (C). Results: Human MM cell lines (HMCLs) and pt samples were cultured at different concentrations (1 vs. 10 vs. 100 cells/microwell), demonstrating a growth advantage w vs w/o M210B4 (a). Liquid overlay technique allowed cluster formation of pt BM cells and a prolonged propagation for up to 20d compared to our 2D models (b). Apoptotic changes were assessed by confocal microscopy of RPMI8226 co-expressing fluorescently labelled histone 2B-mCherry (red, c) and cytochrome c-GFP (green) as indicators of late and early apoptosis. Comparing bone marrow stroma cell lines (BMSCLs) w regard to their MM growth support, human HS-5 proved superior to M210B4 (d). We observed decreased CXCR4 expression w vs. w/o BMSCs, possibly suggesting a dynamic regulation of homing molecules (e). Our model is currently being used to test a combined approach of apcin/proTAME (cyclin B stabilizers) w ARRY-520 (KSP inhibitor). Conclusions: We provide an innovative approach to improve ex vivo compound screening in MM using a 3D high-throughput coculture platform mimicking the BM microenvironment. Our MM specific model may be a suitable tool to better predict in vivo anti MM drug potencies and thus might help to better select drugs and to more reliably estimate the likelihood of their later clinical adoption.

Disclosure: No conflict of interest disclosed.

166

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

dition it impacts on the support function by decreasing the secretion of important growth factors for B-cells. Similar results were observed in first in vivo studies. Taken together our results imply that lenalidomide interrupts an important stromal cell function thereby influencing survival of MM cells. Disclosure: No conflict of interest disclosed.

Freier Vortrag

GIST / Urogenitale Malignome / sonstige Tumore V553

Longitudinal analysis of the impact of the introduction of tyrosine kinase inhibitor therapy on overall survival of patients with gastrointestinal stromal tumors treated at the West German Cancer Center Falkenhorst J.1, Treckmann J.W.2, Nguyen B.-P.3, Podleska L.-E.2, Reis A.-C.4, Virchow I.1, Schuler M.1,5, Bauer S.1,5 Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Innere Klinik (Tumorforschung), Essen, Germany, 2Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Allgemeinchirurgie, Viszeral- und Transplantationschirurgie, Essen, Germany, 3Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Klinik für Gynäkologie und Geburtshilfe, Essen, Germany, 4Universitätsklinikum Essen, Westdeutsches Tumorzentrum, Institut für Pathologie, Essen, Germany, 5Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Essen, Essen, Germany 1

Fig. 1. Synopsis of 3D in vitro trials. Disclosure: No conflict of interest disclosed. V552

Re-educating myeloma associated macrophages with lenalidomide Bruns H. , Gehlen H. , Nolting J. , Pasemann S. , Brossart P. , Mackensen A.1, Gerbitz A.1 1

1

2

1

2

Uniklinikum Erlangen, Dept. of Internal Medicine 5 – Hematology/Oncology, University of Erlangen, Erlangen, Germany, 2Universitätsklinik, Bonn, Germany 1

The bone marrow niche plays a critical role in determining the fate of malignant plasma cells in multiple myeloma (MM). Macrophages are an abundant component of the stromal cell compartment and are believed to support proliferation, survival, and drug resistance of MM cells. Conversely, macrophages can directly kill tumor cells and participate in antitumor immune responses as effector cells. The anti-MM activity of lenalidomide is thought to be mediated, in part, by targeting the stromal support, but its precise influence on the phenotype or the effector functions of macrophages is still unclear. To investigate the effect of lenalidomide on the interaction between macrophages and malignant plasma cells in vitro, we coincubated lenalidomide pretreated generated macrophages with several MM cell lines, and analyzed the viability, proliferation and phenotype. For in vivo studies we utilized 5TMM mice, a suitable animal model for MM. In addition, macrophages in the bone marrow of MM patients treated with lenalidomide were characterized by immunohistochemistry and flow cytometry. We showed, that infiltrating macrophages in the bone marrow of MM patients display an anti-inflammatory M2-like phenotype characterized by the expression of surface marker CD163, CD206, PD-L1 and cytokine/ chemokine secretion (e.g. IL10, CXCL10, APRIL, BAFF and RANKL). Incubation of macrophages with lenalidomide in vitro, substantially changed their transcriptional program (e.g. downregulation of IRF4 and upregulation of IRF5) and their phenotype (e.g. downregulation of the surfaces marker CD163, CD206, and upregulation of CD40 and CD86). Furthermore, we show that lenalidomide treatment decreases the expression of RANKL, BAFF and APRIL, while tumoricidal effector molecules (e.g. TRAIL, cathelicidine, Granzyme B) were increased. When lenalidomide treated macrophages were cocultured with MM cells significant cytotoxicity was detected, for all MM cell lines tested. In contrast, untreated macrophages promote tumor growth and viability of MM cells. Lenalidomide in vitro influences macrophages by reverting an anti-inflammatory M2 like profile to a more immunogenic phenotype. In ad-

Abstracts

Introduction: Gastrointestinal stromal tumors (GIST) are characterized by driver mutations of the receptor tyrosine kinases KIT or PDGFRA. The KIT-inhibitor imatinib (IM) has revolutionized the treatment for patients with both localized and metastatic GIST. Additional advances include improved diagnostics, multimodal treatment and novel KIT inhibitors such as sunitinib (SU) and regorafenib. Very little is known, how these improvements have changed overall survival (OS) over time. Methods: Patients diagnosed with GIST since the time of introduction of IM (2001) were identified from an institutional database (n = 440). We analyzed OS since date of diagnosis, since first treatment with IM for metastatic disease, since IM failure, and since SU failure. For comparison 2 to 4 subgroups were formed by the year of event. Kaplan-Meier (KM) analyses were calculated for each covariate. Results: OS for unselected patients improved over time (2001–2004 vs. 2005–2008 p = 0.038, and 2001–2004 vs. 2009+ p = 0.005) with albeit shorter follow-up and more patients diagnosed at earlier stages in the latter group. Median OS of patients with metastatic GIST has substantially improved (2001–2003 vs. 2004–2008 vs 2008+ : 62 vs 75 months vs not reached) resulting in 5-year-OS-rate of 57 vs. 57 vs. 75% (p = 0,027). 3-year-OS-rate was 42 and 92% when calculated from failure of IM (2001– 2008 vs 2008+). We observed a trend towards better survival for patients failing SU before 2008 vs. since/after 2008 (median OS 14 vs. 21 months, p = 0.051). Conclusions: The survival of patients with GIST consulting a single high volume center has further improved since the introduction of IM in 2001. Contributing factors include improved pathological diagnostics, introduction of adjuvant IM, increased awareness through an active patient advocacy group, systematic follow-up programs, center-based treatment, introduction of second- and third-line treatments. Disclosure: Johanna Falkenhorst: No conflict of interest disclosed. Sebastian Bauer: Advisory Role: Pfizer; Financing of Scientific Research: Novartis, Pfizer, Bayer; Expert Testimony: Novartis.

Oncol Res Treat 2015;38(suppl 5):1–270

167


CONTENTS AUTHOR INDEX

V554

Risk-adapted screening for toxin associated bladder cancer: Results of health service research of IQUO with the risk assessment tool RiskCheck Bladder Cancer© in daily routine work in office based urology Lüdecke G.1, Geiges G.2, König F.3, Interessenverband zur Qualitätssicherung der Arbeit niedergelassener Uro-Onkologen in Deutschland e.V. Universitätsklinikum Gießen und Marburg GmbH, Klinik und Poliklinik für Urologie, Kinderurologie und Andrologie, Giessen, Germany, 2 Urologische Praxis, Lietzenburger Str. 54, Berlin, Germany, 3Urologische Gemeinschaftspraxis, ATURO, Mecklenburgische Str. 27, Berlin, Germany 1

Introduction: The open-access questionnaire RiskCheck Bladder Cancer (RCBC) was proven in daily routine work from German urologists organized in the health services research foundation IQUO on asymptomatic patients to identify BC risk exposure and its relation to detectable tumors. Methods: The RCBC questionnaire resulted in a risk stratification with low- intermediate- and high risk in relation to personal-, smoking-, occupational- and medical risk factors. IBM-SPSS 22 was used for descriptive statistics and effectiveness was proven by classification tree analysis and cross-table analysis with Chi-square test, significance α < 0.05. Results: Out of 359 checked asymptomatic persons 311 (86.6%) were negative for tumor and 48 (13.4%) had a detectable tumor. Male to female was 332 to 27. Smoking was the most dominant risk factor. Non-smoker 52.4%, former smoker 26%, active smoker 21.6%, range of consumed cigarettes 0 to 755,600, consumption classes: < 20,000 5.2%, 20,000 to 100,000 27.9%, 100,000 to 300,000 45.4% and > 300,000 21.6%. According to the ASCO guideline 13.6% were candidates for lung cancer screening based on their epidemiology and smoking behavior. The risk classification showed 62.4% low-risk, 17.3% intermediate-risk and 20.3% high-risk. The RCBC risk assessment showed a significant relation between the exposed risk factors and the detectable bladder cancers (p < 0.01). The classification tree analysis identified age, gender, type of smoking and live time consumption (above 219,000 cigarettes) as relevant factors. Sensitivity 56.3%, specificity 65.3%, NPV 90.6%, PPV 20.0%, false positive rate 43.8%, false negative rate 34.7%, accuracy 64.1%. Risk-adapted screening resulted in an increase of effectiveness of 214.8 compared to mass screening. Conclusion: Preventive medical care becomes effective by RCBC. Via the secure VPN based questionnaire handling the IQUO can demonstrate that this tool can work in the hands of urologists. A yearly recall control in urological offices for the risk population (32.3%) can be organized. The assessment is work effective, aim achieving and in result cost effective. The questionnaire RCBC integrates evidence based bladder cancer inductors, is easy in use and as a open-access tool available in 10 languages via internet to all medical services. www.riskcheck-bladder-cancer.info. Furthermore RCBC is transferred to the iOS platform as RCBC2 including bladder- and lung cancer screening and available as an iPad APP. Disclosure: No conflict of interest disclosed. V555

A mutation in Exon 28 of TSC2 [TSC2R1125W] predicts sensitivity of a heavily pretreated metastatic germ cell tumor to temsirolimus Maerz W.1, Schröder C.2, Haj Abdo K.1, Fischer S.3, Riess O.2, Blumstein N.1 Paracelsus Klinik, Osnabrück, Germany, 2Genes&Therapy GmbH, Tübingen, Germany, 3Klinikum, Ibbenbüren, Germany 1

Background: Contemporary genomic analysis of tumor tissue may have an impact on the choice and success of treatment in difficult to treat solid tumors. Commercially available tumor gene panels generate a wealth of data, while upfront evidence of druggability of detected mutations may be lacking. Case report: Here we describe a patient with a 10 year history of metastatic germ cell tumor dedifferentiated into sarcomatoid subtype with persistent metastatic disease to lung and mediastinum. 6 different lines

168

Oncol Res Treat 2015;38(suppl 5):1–270

of chemotherapy – incorporating high dose consolidation chemotherapy with autologous stem cell support on 2 occasions – were unable to induce a long lasting remission. The patient remained sensitive to a 7th line of chemotherapy with adriamycin and ifosfamide until this had to be stopped due to anticipated cumulative anthracyclin cardiotoxicity. Subsequent mediastinal tumor progress called for action. A surgical tissue biopsy was obtained and submitted to tumor gene panel analysis. [Genes & Therapy GmbH Tuebingen Germany somatic_cancer_panel_v2]. Mutations in several potentially relevant genes were detected, including v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 [ERBB2L632V], tuberous sclerosis complex 2 [TSC2 R1125W] and mechanistic target of rapamycin [MTORR769C]. ERBB2-immunohistochemistry however did not show membrane bound overexpression. The detected 2 mutations along the MTOR pathway paired with anecdotal published evidence (N Engl J Med 2014; 371:1426–1433) suggested MTOR-blockade as a therapeutic option. Treatment with temsirolimus (25 mg i.v. weekly) resulted in good partial remission after 2 weeks and complete remission after 8 weeks. Subsequent temsirolimus maintenance (25 mg i.v. q21 days) resulted in tumor progression 3 months later. At this time a repeat biopsy was performed before intensified retreatment with MTOR-blocking agents. Unfortunately at this time oral Everolimus 10 mg daily for two weeks and subsequent Temsirolimus 25 mg i.v weekly for three weeks were no better than maintaining a stable disease. Conclusion: Tumor gene panel analysis inspired empirical therapy with Temsirolimus, helped to outperform seven prior lines of chemotherapy in this patient and generated preliminary evidence of the TSC2R1125W mutation as a predictive marker of response to MTOR-blockade. Dose intensification and reanalysis of tissue obtained after 1st progression on Temsirolimus maintenance is underway and will be reported at the meeting. Disclosure: Wolfgang Maerz: Other Financial Relationships: Novartis : travel grants. Norbert Blumstein: No conflict of interest disclosed. V556

Molecular profiling of cisplatin-resistant testicular germ cell tumors Oing C.1, Bokemeyer C.1, Russell K.2, Millis S.Z.3, Bender R.3, Gatalica Z.3, Voss A.2 University Medical Center Hamburg-Eppendorf, Oncology, Hematology and BMT with the section of Pneumology, Hamburg, Germany, 2Caris Life Sciences, Basel, Switzerland, 3Caris Life Sciences, Phoenix, United States 1

Introduction: Testicular germ cell tumors (TGCT) are the most common malignancy among men under the age of 40 years and the incidence has steadily been rising over the last decades. Since the introduction of cisplatin-based combination chemotherapy in the late 1970ies, GCTs have become a highly curable disease with cure rates of more than 90%. Failing cisplatin-based chemotherapy confers a dismal outcome and treatment options are limited. Personalizing treatment upon molecular diagnostic tools to identify targetable lesions and to predict response to targeting agents may offer individual treatment options after failure of standard chemotherapy. Methods: Samples from 50 advanced stage TGCT patients failing standard treatment referred to Caris Life Sciences between 2009 and February 2015 were analyzed. Diagnoses were collected from referring physicians and classified based-on pathology reports and clinical history. Analyses were performed per physician request and comprised a combination of protein expression (immunohistochemistry), genomic sequencing (Sanger, NGS or pyrosequencing), gene amplification (FISH or CISH), and/or RNA fragment analyses. Results: In this group of treatment-resistant TGCTs, the most commonly gene expressional aberrations included upregulation of TOPO2A (89%) and several drug-resistance associated proteins, i.e. ABCG2, MRP1, MDR-1 and TLE3 in 75%, 68%, 21% and 41%, respectively. Proteins commonly downregulated were ERCC1, MGMT, PTEN and RRM1 in 81%,

Abstracts


CONTENTS AUTHOR INDEX

67%, 61%, and 61%, respectively. Amplifications of the EGFR, HER2, and cMET gene were found in 21%, 4%, and 9%, and a single patient analyzed showed a PIK3CA amplification. In 14 samples analyzed by NGS only few mutations in the KRAS (7%), cKIT (14%), EGFR (7%), SMAD4 (7%) and TP53 (31%) gene were detected. Pyrosequencing revealed one more KRAS mutation. No other RAS-subtype mutations were found. Oncogenic pathway profiling revealed almost all patients to be MAPK and mTOR wild-type except for PTEN expression loss and a PIK3CA amplification. MAPK activation only occurred through KRAS gene mutations. Conclusions: Mutations of well-known oncogenic drivers and tumor suppressors in treatment-resistant TGCTs are scarce. However, treatment-resistance associated proteins are often dysregulated. Predictive biomarkers may be established to guide individual treatment decisions, e.g. to target cKIT, HER2 or cMET in single patients. Disclosure: Christoph Oing: No conflict of interest disclosed. Andreas Voss: Employment or Leadership Position: Vice President of Medical and Clinical Affairs for Caris Life Sciences in Europe.

increased DNA repair ability, compared to their wildtype counterparts. We propose that it is this enhanced genome maintenance capacity that prevents malignant transformation and promotes oncogene resistance. Disclosure: No conflict of interest disclosed.

Freier Vortrag Immuntherapie II V559

Leukemic stem cells are immunogenic targets for cytotoxic CD8-positive T cells Greiner J.1,2, Zhang L.1,3, Rojewski M.4, Fekete N.4, Erle A.4, Bullinger L.1, Hofmann S.1, Götz M.1, Döhner K.1, Ihme S.5, Döhner H.1, Buske C.5, Schneider V.1 University of Ulm, Department of Internal Medicine III, Ulm, Germany, Diakonie Hospital Stuttgart, Department of Internal Medicine, Stuttgart, Germany, 3Tongji Hospital, Department of Oncology, Wuhan, China, 4University of Ulm, Institute of Clinical Transfusion Medicine, Ulm, Germany, 5University of Ulm, Institute of Experimental Cancer Research & Department of Internal Medicine III, Ulm, Germany 1 2

V557

Combined Chk1- and MK2-inhibition displays robust synergy in KRAS-mutant cancer Dietlein F.1, Kalb B.1, Jokic M.1, Torgovnick A.1, Schmitt A.1, Büttner R.1, Thomas R.1, Reinhardt H.C.2 Uniklinik, Köln, Germany, 2Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany 1

KRAS is one of the most frequently mutated oncogenes in human cancer. Despite substantial efforts, no clinically applicable strategy has yet been developed to effectively treat KRAS-mutant tumors. We performed a cell line-based screen and identified strong synergistic interactions between cell cycle checkpoint-abrogating Chk1- and MK2 inhibitors, specifically in KRAS- and BRAF-mutant cells. We further show that KRAS-mutant cancer displays intrinsic genotoxic stress, leading to tonic Chk1- and MK2 activity. We demonstrate that combined Chk1- and MK2 inhibition leads to mitotic catastrophe in KRAS-mutant cells. This actionable synergistic interaction is extensively validated in vivo using xenograft models, as well as distinct Kras- or Braf-driven autochthonous murine cancer models. Lastly, we show that combined checkpoint inhibition induces apoptotic cell death in KRAS- or BRAF-mutant tumor cells directly isolated from patients. These results strongly recommend simultaneous Chk1- and MK2 inhibition as a therapeutic strategy for the treatment of KRAS- or BRAF-driven cancers. Disclosure: No conflict of interest disclosed. V558

Cdkn1aSUPER mice display an increased DNA repair capacity and are protected from mailgnant transformation Torgovnick A.1, Heger M.1, Reinhardt H.C.1 Uniklinik, Köln, Germany

1

CDKN1A is an important target gene within the p53 signaling network, which encodes for the CDK inhibitor p21. To experimentally approach the duality of the p53 response, which can lead to the induction of cell death, through the transactivation of pro-apoptotic target genes, and to the induction of a cell cycle arrest, through the transactivation of cell cycle-regulating target genes, we generated a Cdkn1a transgenic mouse. This mouse carries a conditional extra copy of Cdkn1a. We show that our novel strain carries a single extra copy of Cdkn1a, which is expressed in a p53- and DNA damage-specific manner. This increased Cdkn1a gene dosage leads to a remarkable protection from transformation in vivo. We specifically use a DMBA-TPA-induced papilloma model, a Kras-driven lung adenocarcinoma model and a 3MC-induced sarcoma model to unequivocally show that one extra copy of Cdkn1a provides robust tumor protection, similar to what we observe in mice carrying an extra copy of Tp53. Mechanistically, we show that Cdkn1a transgenic mice display an

Abstracts

To prevent relapse after intensive chemotherapy and/or allogeneic stem cell transplantation is one of the major challenges in treatment of patients with acute myeloid Leukemia (AML). Leukemic stem cells (LSC) might be the source for leukemic disease self-renewal and account for disease relapse after treatment which makes them a critical target for therapeutic options. Immunotherapeutic approaches might overcome the tumor-escape mechanisms of LSC. Leukemia-associated antigens (LAAs) represent antigens that are recognized by cytotoxic T-lymphocytes (CTL) and are such target structures for specific immunotherapy. We compared LAA expression in enriched CD34+CD38- AML fraction (called here LSC-EF) to that of enriched HSC of healthy donors and AML bulk cells. Moreover, we investigated the expression patterns of co-stimulatory molecules of LSC, bulk AML cells and enriched HSC. Expression levels of LAA detected via microarrays were confirmed by quantitative RT-PCR. Functional immunoassays were performed to examine targeting of AML progenitors by cytotoxic T cells (CTL) using colony forming assays (CFUs). Furthermore, in a xenotransplant model we demonstrated that AML stem cells can partly be controlled by peptide-stimulated CTL. Several LAA were highly expressed in in LSC-EF, a prerequisite for immunotherapeutic approaches against LSC. For example, PRAME, RHAMM, WT1 and Proteinase 3 showed significant higher expression in LSC-EF compared to normal HSC whereas Survivin, HAGE, SSXIP2 and Aurorakinase A did not indicate a significant differential expression. The LAAs PRAME, RHAMM and WT1 are even higher expressed in LSC-EF than in leukemic bulk qualified these LAAs as interesting potential targets. In contrast, Proteinase 3 indicates a higher expression on leukemic bulk but lower in LSC-EF. RT-PCR confirmed our array data. In our immunoassays, T cells stimulated against various LAA indicated a significant inhibition of CFUs in AML patient samples. The LAA PRAME, RHAMM and WT1 showed highest immunogenic responses with a range up to 58–83%. In a proof of principle xenotransplant mouse model, PRAME-stimulated CTL targeted AML stem cells, reflected by a delayed engraftment of leukemia (p = 0.0159) Taken together, we demonstrated the expression of several LAA in LSC. LAA-specific T cells are able to hamper LSC in immunoassays and in a mouse model, which suggests that immunotherapeutic approaches have the potential to target malignant stem cells. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

169


CONTENTS AUTHOR INDEX

V560

In vitro and in vivo imaging of the migratory behavior of tumorantigen-specific CD40-activated B cells in the setting of B cell-based cancer immunotherapy Wennhold K.1,2, Thelen M.1,2, Shimabukuro-Vornhagen A.1,2, von Bergwelt-Baildon M.1,2 Uniklinik Köln, Interventionelle Immunologie, Köln, Germany, 2Uniklinik Köln, Innere Medizin I, Köln, Germany 1

Efficient antigen presentation is a prerequisite for the development of a T-cell mediated immune response. Dendritic cells (DCs) are known to be the most prominent professional antigen-presenting cells (APCs). However, they have several disadvantages as cellular adjuvant in cancer immunotherapy. Therefore, an alternative approach was developed, in which polyclonal B cells can serve as potent APCs by treatment with the CD40 ligand. We demonstrated that CD40-activation dramatically improves antigen presentation by B cells, efficiently inducing naive and memory CD4+ and CD8+ T-cell responses. Contrary to DCs, we know little about the migration behavior of B cells in vivo. Therefore, we investigated the interactions between CD40-activated B (CD40B) cells and cytotoxic T cells in vitro and the migration behavior of CD40B cells in vivo. Dynamic interactions of human antigen-presenting cells (APCs) and T cells were observed by time-lapse videomicroscopy. The migratory and chemoattractant potential of murine CD40B cells was analyzed by flow cytometry and by standard transwell migration assays. GFP+ or luciferase+ CD40B cells were used for subsequent in vivo studies. Murine CD40B cells show migratory features similar to human CD40B cells. They express important lymph node homing receptors; these are functional and induce chemotaxis of T cells in vitro. Striking differences were observed with regard to interactions of human APCs with T cells. CD40B cells differ from DCs by displaying a rapid migratory pattern undergoing highly dynamic, shortlived and sequential interactions with T cells. In vivo, CD40B cells home to the secondary lymphoid organs, where they accumulate in the B cell zone before traveling to the B/ T cell boundary. Excitingly, tumorantigen-specific CD40B cells, which are highly potent APCs, migrate to the tumor site in tumor-bearing mice. Taken together, these data show that CD40B cells home to secondary lymphoid organs and to the tumor and physically interact with T cells, thus underscoring their potential as cellular adjuvant for cancer immunotherapy. Disclosure: Kerstin Wennhold: No conflict of interest disclosed. Michael von Bergwelt-Baildon: Advisory Role: MSD SHARP & DOHME GMBH, Astellas Pharma GmbH, Roche, Amgen; Financing of Scientific Research: MSD SHARP & DOHME GMBH, Astellas Pharma GmbH, Roche, Amgen; Expert Testimony: MSD SHARP & DOHME GMBH, Astellas Pharma GmbH, Roche, Amgen. V561

Boosting ADCC activity of therapeutic antibodies by concomitant stimulation of FcγRIIIa and activating NK cell receptors Kellner C.1, Klausz K.1, Kahrs A.-K.1, Oh J.1, Humpe A.1, Parren P.W.H.I.2,3, van de Winkel J.G.J.2, Gramatzki M.1, Peipp M.1 Division of Stem Cell Transplantation and Immunotherapy, Kiel, Germany, Genmab, Utrecht, Netherlands, 3Dept. of Cancer and Inflammation Research, Odense, Denmark 1 2

Background: Strategies boosting distinct effector mechanisms mediated by therapeutic antibodies represent promising approaches to enhance antibody-based cancer immunotherapy. Here, recombinant immunoligands, which engage the stimulatory NK cell receptors natural killer group 2 member D (NKG2D) or NKp30 were analyzed for their capacity to enhance antibody-dependent cell-mediated cytotoxicity (ADCC), a major mechanism of action triggered by therapeutic antibodies. Methods: The NKp30-specific ligand B7-H6 or the NKG2D-specific ligand ULBP2, were fused to scFv fragments directed against tumor-associated antigens CD20 or HER2. The purified fusion proteins specifically bound to antigen-positive tumor cells and simultaneously reacted with

170

Oncol Res Treat 2015;38(suppl 5):1–270

the corresponding NK cell receptors, as shown by flow cytometry. The cytolytic capacity of the immunoligands when applied in combination with monoclonal antibodies (lymphoma model: rituximab, daratumumab; solid tumor model: trastuzumab, cetuximab) were investigated by chromium release assays with cell lines, or with freshly isolated tumor cells employing allogeneic or autologous NK cells. Results: The CD20-specific immunoligands ULBP2:CD20-scFv and B7-H6:CD20-scFv triggered lysis of tumor cells and had the capacity to augment ADCC, with ULBP2:CD20-scFv being more potent than B7H6:CD20-scFv. ULBP2:CD20-scFv in particular was efficacious in boosting ADCC when lymphoma cells from patients were employed as target cells. Enhanced lysis was also achieved when autologous, patient-derived NK cells were analyzed. The ADCC enhancing capacity of recombinant immunoligands was confirmed with HER2-specific constructs, ULBP2:HER2-scFv and B7-H6:HER2-scFv. Both constructs enhanced ADCC by the therapeutic antibodies trastuzumab and cetuximab synergistically. In contrast, almost no improvements were obtained when different IgG antibodies (e.g. cetuximab and trastuzumab) were combined, emphasizing the benefits of approaches that allow co-engagement of two different NK cell receptors. Conclusions: Triggering activating NK cell receptors NKG2D or NKp30 by immunoligands in parallel to FcγRIIIa boosts ADCC by therapeutic antibodies synergistically and may represent a general approach to improve antibody-based cancer immunotherapy. Based on these findings, a Dual-Dual Targeting Concept (DDT-Concept) addressing two target antigens and two triggering receptors may be realized, allowing specific amplification of NK-cell responses at the tumor site. Disclosure: No conflict of interest disclosed. V562

Evaluating human T-cell therapies of systemic cytomegalovirus organ disease in HLA-transgenic mice Thomas S.1, Lemmermann N.2, Klobuch S.1, Podlech J.2, Plachter B.2, Theobald M.3, Reddehase M.2, Herr W.1 Universitätsklinikum Regensburg, Innere Medizin III, Hämatologie und Onkologie, Regensburg, Germany, 2Universitätsmedizin Mainz, Inistitut für Virologie, Mainz, Germany, 3Universitätsmedizin Mainz, Innere Medizin III, Mainz, Germany 1

Reactivation of human cytomegalovirus (HCMV) can cause severe disease in recipients of hematopoietic stem cell transplantation. Although preclinical research in murine models as well as clinical trials have provided ´proof of concept´ for infection control by pre-emptive CD8 T-cell immunotherapy, there exists no predictive model to experimentally evaluate parameters that determine antiviral efficacy of human T cells in terms of virus control in functional organs, prevention of organ disease, and host survival benefit. Here we introduce a novel mouse model for testing HCMV epitope-specific human T cells. In this, we combined the well-described murine model of mCMV infection of the immunocompromised host with the strong T-cell immunogenicity of the HLA-A*0201 restricted HCMV epitope pp65495–503 NLVPMVATV (NLV). We generated a chimeric recombinant mCMV expressing the NLV epitope (mCMV-NLV) during the infectious cycle to allow organ manifestation of the infection in the natural host similar to that seen in immunocompromised patients. After infection of HLA-A2.1 transgenic immunocompromised NOD/SCID/ IL-2rg–/– (NSG/HHD) mice, mCMV-NLV resulted in a rapid systemic infection that could be effectively combated by adoptively transferred human NLV-specific CD8 T cells as well as by human CD8/CD4 T cells transduced with an NLV-specific T-cell receptor. Upon transfer, the T cells infiltrated host tissues in an epitope-specific manner, confining the infection to nodular inflammatory foci. This resulted in a significant reduction of viral load, prevention of organ pathology, and prolonged survival. Collectively, these data demonstrated that NLV-specific human T cells mediate HCMV NLV-specific immune control in mCMV-NLV infected NSG/ HHD mice, even across the human-mouse species barrier.

Abstracts


CONTENTS AUTHOR INDEX

The model has thus proven its potential for a preclinical testing of the protective antiviral efficacy of HCMV epitope-specific human T cells in the evaluation of new approaches to an immunotherapy of CMV disease.

V564

Peptide-based immunotherapy for B cell lymphomas based on the recurrent oncogenic MYD88 L265P mutation

Disclosure: No conflict of interest disclosed.

Nelde A.1,2, Stickel J.S.1, Kowalewski D.J.2, Kanz L.1, Langerak A.W.3, Muggen A.F.3, Fend F.4, Rammensee H.-G.2, Stevanovic S.2, Weber A.N.R.2

V563

1

Novel human IL-15 superagonist promotes long-term persistence and reactivity of EBV- and leukemia-specific CD8+ T lymphocytes with stem cell memory and central memory properties in humanized mice Mades A.1, Khan S.A.1, Theobald M.1, Hartwig U.F.1 III. Medizinische Klinik, Universitätsmedizin der Johannes GutenbergUniversität Mainz, Hämatologie, Internistische Onkologie & Pneumologie, Mainz, Germany 1

Background: Adoptive cellular therapy using leukemia-reactive T cells has evolved as a promising strategy to improve antileukemic immunity. Moreover, leukemia-reactive stem cell memory (TSCM) or central memory (TCM) T cells as well as IL15supplementation have been shown to improve clinical responses by promoting T cell homeostasis, function and the formation of T cell memory. We therefore investigated whether a novel IL-15 superagonist could augment engraftment, persistence and cytolytic potential of human CD8+ T cells in humanized NSG mice. Methods: The novel IL-15 superagonist, referred to as ILR-Fc was generated by linking human IL-15N72D, a high affinity IL-15 point mutant to the IL-15Rα-Sushi domain and an IgG1-Fc domain to increase its affinity to the physiological IL15Rβγ-complex and prolong bioavailability. HLA-A2-, Epstein-Barr Virus- (EBV)- and acute myeloid leukemia(AML)- reactive CD8+ T cells were generated from naïve CD45RA+CD8+ T cells by repetitive stimulation using either HLA-A2 expressing K562 transfectants, autologous EBV-peptide loaded DCs or HLA-matched primary AML blasts, respectively. T cell cultures were additionally supplied with the GSK3 inhibitor TWS119 that facilitates the generation of antigen-specific T cells with a TSCM and TCM phenotype. Results: The Expi293 Expression System (Invitrogen) and protein-A affinity based purification were applied to produce soluble ILR-Fc with yields of ≥1 mg recombinant protein/25 ml culture. In vitro bioactivity analyses revealed that ILR-Fc was ≥10 fold more active on M07e cells than recombinant IL-15 produced in E. coli. Strong effects on expansion of CD8+ T cells could be observed following injection of IL-Fc into wild type C57BL/6 mice and humanized NSG mice following reconstitution with CD34+ hematopoietic stem cells. Moreover, upon transfer of EBVand AML-reactive CD8+ CTLSCM/CM into NSG mice engrafted with autologous EBV-immortalized B cells or HLA-matched AML blasts, we observed prolonged persistence and superior antitumor-reactivity in mice receiving weekly i.p. injections of 50–60 ug ILR-Fc/mouse. This extended bioactivity was partially attributed to an increased half-life of ILR-Fc in the serum as determined by IL-15 ELISA. Conclusions: These studies demonstrate that ILR-Fc has potent and prolonged bioactivity to facilitate CD8+ T cell persistence and activity in lymphopenic hosts and might thus serve as a valuable tool to improve selective CD8+ T cell-mediated immunotherapy.

University Hospital Tübingen, Department of Hematology and Oncology, Tübingen, Germany, 2Interfaculty Institute of Cell Biology, Department of Immunology, Tübingen, Germany, 3Erasmus MC Rotterdam, Department of Immunology, Rotterdam, Netherlands, 4University Hospital Tübingen, Department of Pathology, Tübingen, Germany

Non-Hodgkin Lymphomas (NHL) are frequent malignancies with considerable mortality. A recurring somatic and oncogenic driver mutation in the Toll-like receptor (TLR) adaptor gene MYD88, Leu 265 Pro (L265P) was identified in up to 90% of certain NHL subtypes. Because MyD88 L265P is a widely occurring and tumor-specific mutation, we investigated the suitability of L265P-containing peptides for CD8 T cell mediated immunotherapy as a new therapeutic approach for L265P+ NHL. Based on in silico predictions we identified potential HLA ligands encompassing the L265P mutation for several HLA class I restrictions. Functional analysis of the HLA class I L265P-derived NHL-specific ligands with regard to induction of T cell responses identified a set of peptides immunogenic for HLA B*07 and B*15. Memory T cell responses were detected in a L265P+ B*15 CLL patient and efficient T cell priming was demonstrated in vitro using naïve T cells of healthy volunteers (HVs). In detail, three B*07 and one B*15 restricted peptides were analyzed using artificial antigen-presenting cells (aAPCs) in in vitro priming experiments in three to six HVs, respectively. For all tested peptides a proliferation of peptide-specific CD8+ T cells could be detected after in vitro priming. For the B*07 restricted peptides peptide-specific CD8 T cells could be induced in 6/6 (100%), 1/3 (33%) and 3/4 (75%) HVs, respectively, with a maximum frequency of 14.1% within the CD8+ T cell population. For the B*15 restricted peptide peptide-specific T cells could be induced in 2/3 (66%) HVs with a maximum frequency of 9.5% tetramer-positive cells within the CD8+ T cell population. The functionality and specificity of peptide-specific T cells was validated by intracellular staining for IFNγ and TNFα as well as for the expression of the degranulation marker CD107a. In 2/2 (100%) donors we detected specific and functional CD8+ T cell populations after stimulation with the mutated peptide, but not after stimulation with the corresponding wild type peptide. Furthermore, the peptide-specific cytotoxic activity of CD8+ T cells was demonstrated in a VITAL assay. The polyclonal T cells lysed autologous PBMCs loaded with the mutated peptide (11.4% specific lysis at an effector to target ratio of 0.7 to 1), but not cells presenting the wild type peptide. These data highlight MYD88 mutation-specific peptide-based immunotherapy as a novel personalized new treatment approach for patients with L265P+ NHL. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

171


CONTENTS AUTHOR INDEX

Freier Vortrag

V566

Patientenberatung und Krebsregister

Which advices need adolescents and young adults after stem cell transplantation?

V565

Pulewka K.1, Hebestreit N.2, Strauß B.1, Hochhaus A.3, Hilgendorf I.3

Psycho-oncologic interventions for parents of cancer patients: Systematic review Koehler M.1, Hoppe S.1, Peplinski D.1, Richter D.2, Frommer J.3, Flechtner H.-H.4, Fischer T.1, Studiengruppe AYA-Netzwerk University Medical Center Magdeburg, Department of Hematology and Oncology, Magdeburg, Germany, 2University Medical Center Leipzig, Department of Medical Psychology and Medical Sociology, Leipzig, Germany, 3 University Medical Center Magdeburg, Department of Psychosomatic Medicine and Psychotherapy, Magdeburg, Germany, 4University Medical Center Magdeburg, Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatic Medicine, Magdeburg, Germany 1

Introduction: How to deal with the psychological burden of parents of cancer patients within the oncological treatment process often represents a challenge for all the parties involved. The current study is purposed to answer the research question: “Are there evaluated psycho-oncologic interventions concerning the psychosocial needs of parents of cancer patients?” Methods: Literature databases and clinical trial registries were systematically searched for randomized controlled trials (RTCs), which compare the effects of an intervention focused on the psychosocial needs of parents of cancer patients of any age with a control condition. The selection of clinical trials was conducted according to PICOS criteria. As primary outcome variable we defined psychosocial health needs which were operationalized on the basis of domains named mental health, coping strategies and supportive care needs (SCN). The study is compiled according to the DARE-criteria, the PRISMA statement and is registered at PROSPERO (International prospective register of systematic reviews: CRD42014007401). Results: We selected 12 RTCs comparing intervention vs. control condition which included 1848 Parents (82% mothers) and 1520 patients (mean age 8.6 years, 55% systemic cancer). The median treatment included 5 sessions which were conducted over a period from 1 day (4 sessions) up to 6 month (8 sessions of 90 minutes, every 3 weeks). The most commonly used format was skill training in individual sessions. All interventions were based on a cognitive-behavioral approach. The primary outcome variables were assessed only in two domains: mental health and coping strategies. Conclusion: Future clinical studies should consider multiple missing aspects (e.g. homogenous samples regarding the tumor entities). Particularly the parents of adolescent and young adults (AYA) with cancer are still excluded, although it is not an exclusive pediatric challenge, to perceive the specific needs of parents in their role as family caregivers for their “children” suffering from cancer. Future interventions should strengthen the use of psychodynamic-supportive treatment techniques and include more parent specific contents: In our view the verbalization of subjective ideas of responsibility is particularly important as well as to consider, that the parent-patient-relation can be influenced by drastic changes of the outer and inner reality (e.g., parents’ suffer from adult children’s inclination to regression). Disclosure: No conflict of interest disclosed.

Universitätsklinikum Jena / Institut für Psychosoziale Medizin und Psychotherapie, Jena, Germany, 2Universitätsklinikum Jena / Pflegedirektion, Jena, Germany, 3Universitätsklinikum Jena / Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Jena, Germany 1

Introduction: The potency of psychosocial interventions to adapt on life threatening diseases is well characterized. Adolescent and young adults (AYA) present a minority in the population of stem cell transplanted (SCT) patients, but may have increased consulting requirements, due to long lasting therapeutic interventions falling in a sensible period of development. Thus, AYAs may profit from psychosocial and educational interventions. We therefore sought to evaluate their consulting requirements in a single center survey. Methods: All 33 AYA alive after SCT between 2010 and 2015 and a control group of 32 senior patients transplanted in 2014 at our center were invited to participate. A questionnaire addressing the individual psychosocial and medical consulting requirements was sent to all patients. Results: 39 of 65 questionnaires (60%; 16/33 AYA and 23/32 non-AYAs) were completed and returned. Median age of participating AYA was 33.5 [29–38] years and 55.0 [49–59] years in the control group. Although not significantly different, AYAs predominantly indicated advice in medical (66.7% vs. 47.4% non-AYAs, p = 0.29) and psycho-oncological issues (61.5% vs. 36.8% non-AYAs, p = 0.17), whereas senior patients quoted needs in social legislative queries (76.2% vs. 50% AYA, p = 0.13). On a one to six graded Likert scale issues regarding body shape as well as sexuality were of significantly more concern in AYA compared to controls (median values: 4.5 [4–5] vs. 3 [1–5], p = 0.012). Other aspects, including nutrition, vaccination, medication, alternative medicine, family/relationship, fatigue, skin alterations/alopecia, recreation techniques, follow-up/rehabilitation as well as level of care/premature pension were of high concern in most patients (in median graded 3 to 5), but comparable between both groups. 82.9% of all participating patients would prefer individual consultation instead of grouped information sessions. The majority of patients favour explanatory booklets (66.7%) and internet proposals (71.8%), rather than patient fora (48.7%) as source of information. Conclusions: Both patient cohorts indicated need for advice after SCT by individual consultation. However, AYA reported on increased need for counseling in medical and psycho-oncological issues. They seem to benefit from special themes, i.e. body shape and sexuality. Prospective multicenter studies in this minority of patients are required in order to evaluate the special needs of AYA in the setting of SCT. Disclosure: No conflict of interest disclosed. V567

Concept, implementation and previous experience of a project from the “Bayerische Krebsgesellschaft e.V.” (BKG): Special consultation hour for patients suffering from cancerrelated fatigue Fischer I.1,2, Petsch S.3, Rüffer J.U.2, Heim M.E.2,4, Bojko P.5, Riedner C.6, Rinas N.7, Milani V.8, Schneider E.9, Besseler M.10 Institut für Tumor-Fatigue-Forschung, Emskirchen, Germany, 2Deutsche Fatigue Gesellschaft, Köln, Germany, 3Tumorzentrum der Universität ErlangenNürnberg, Erlangen, Germany, 4Klinik Sokrates, CH-Güttingen, Switzerland, 5 Onkologie Elisenhof, München, Germany, 6Krebsberatungsstelle am Tumorzentrum München in Kooperation mit der Bayerischen Krebsgesellschaft e.V., München, Germany, 7Paracelsus-Klinik, Scheidegg, Germany, 8 Facharztzentrum, Fürstenfeldbruck, Germany, 9Praxis für Psychosomatische Medizin und Psychotherapie, Psychoonkologie, Traumatherapie, Wangen, Germany, 10Bayerische Krebsgesellschaft e.V., München, Germany 1

Cancer-related fatigue (CRF) is defined as a persistent, subjective sense of exhaustion related to cancer or cancer treatment. It can significantly impair normal physical and psychological functioning and, as a consequence

172

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

of this, also every-day life and quality of life. Although many patients are affected and although evidence-based treatment options are available, there is a lack of qualified counseling institutions in Germany. As to date there is no ICD-code for cancer-related fatigue; the care gap can most suitably be closed by a nonprofit organization such as the “Bayerische Krebsgesellschaft e.V.”. Thus, together with the “Institut für Tumor-Fatigue-Forschung”, the BKG has started with the development of a health care structure in Bavaria by offering free-of-cost consultation hours for patients suffering from cancer-related fatigue. The consultation hours are comprised of diagnosis, individual consultancy, education and the offer to take part in training courses which are appropriate for patients with cancer-related fatigue and are orientated on the current available evidence-based treatment options. The first consultation hour took place in September, 2013, in the psycho-social counseling center of the BKG in Nuremberg. Because it has been accepted by patients and because evaluation showed a high degree of patient´s satisfaction, it is now offered in identical form also in four other Bavarian cities. The new consultation sessions are carried out by physicians who have extensive experience both in oncology and psycho-oncology. All consultation sessions will be continuously evaluated in regard to concept and benefit. If experience shows that the treatment has been beneficial, it is planned to offer cancer-fatigue consultation hours from 2016 additionally in further locations of the BKG. The project will be performed in co-operation with the “Deutsche Fatigue Gesellschaft” (DFaG, Köln) and the “Tumorzentrum der Universität Erlangen-Nürnberg”. Concept, implementation and previous experience of the consultation hours will be presented, hoping that cancer organizations in other (federal) states will join us, so that on a medium to long-term basis, all patients with cancer-related fatigue can be adequately treated close to their homes. For this purpose, we are happy to offer our support. Disclosure: No conflict of interest disclosed. V568

“Job perspective” – Development of a MBOR module for oncologic rehabilitation by an interdisciplinary team Leibbrand B.1,2, Exner A.-K.2,3, Kähnert H.2 Salzetalklinik, Onkologie, Bad Salzuflen, Germany, 2Institut für Rehabilitationsforschung Norderney, Bad Salzuflen, Germany, 3Universität, Bielefeld, Germany 1

Introduction: The rehabilitation follows the aim of restoring occupational functioning and preventing premature retirement. Therefore, work-orientated therapies should be an integrated part of rehabilitation (MBOR). The study aim was to design work-orientated therapies (MBOR module “job perspective”) for oncologic rehabilitation and check them by formative evaluation. Methods: The development of “job perspective” was carried out in the rehabilitation team. After testing phase, the module was implemented in the routine care. For evaluation of the module, group interviews with patients and expert interviews with therapists and physicians were conducted. The interviews were evaluated by “category-led” text analysis (Mayring, 2010). In addition, an evaluation of the module was made by questionnaires for patients before (CC: control group, n = 115) and after (IG: intervention group, n = 130) introduction. Results: The module “job perspective” offers work-related therapies especially for office workers and cleaning personnel and other professions. The evaluation showed that the development of the module in the rehabilitation team was positive. It furthered team communication and showed synergy effects. The evaluation of the group interviews with the patients showed that the module was considered practical. The assessment of work-orientated therapies was valuated positively after introduction of the module of IG (60%) but not of CG (11%) (Chi2 = 55.3, p < 0.001). The procedures of addressing work-related issues were evaluated “good” to “very good” in 84% in IG but only 37% in CG (Chi2 = 62.3, p < 0.001). In addition, 69% of IG mentioned that own desires were considered for

Abstracts

future job planning but only 38% in CG (Chi2 = 30.1, p < 0.001). At least 62% of IG would recommend work-related therapies to friends but only 26% of CG (Chi2 = 57.8, p < 0.001). All in all 86% of the IG but only 39% of CG evaluated their rehabilitation positively for integration into employment (Chi2 = 56.9, p < 0.001). Conclusions: The reintegration of cancer patients into working life is a major challenge in rehabilitation. Therefore, the MBOR module “job perspective” was created with an interdisciplinary rehabilitation team. The results emphasize that patients appreciate the consideration of work-related issues in their rehabilitation process. In addition, the results show the importance of interdisciplinary teamwork in the development of new concepts and therapy modules for rehabilitation. Disclosure: No conflict of interest disclosed. V569

CNS tumours in cancer registry of Baden-Wuerttemberg: Handling of missing, incomplete and contradictory data Bezold K.1, Adzersen K.-H.1, Friedrich S.1, Hermann S.1, Becker N.1 DKFZ, Epidemiologisches Krebsregister Baden-Württemberg, Heidelberg, Germany 1

Introduction: Epidemiological cancer registries often suffer from missing, incomplete or contradictory data from different institutions (tumour centres, hospitals, registered physicians and pathologists). The Cancer registry of Baden-Wuerttemberg holds 10.626 reports of CNS tumours. Aim of this work is to suggest a practical best-of algorithm to improve completeness and quality of the registry data. Methods: The data set of each registered CNS tumour is examined for multiple, incomplete, incompatible and/or missing data in respect to morphology, topography and diagnosis. In case of two equal valued morphologies an overarching histology is chosen, e. g. Astrocytoma, NOS for two different morphological astrocytic codes. In case of a missing histology an ICD-O-3 morphology digit is chosen which comes closest to the clinical ICD-10 diagnosis. If histology and ICD-10 diagnosis are incompatible the morphology overrides the ICD-10. In case of two differing topography codes and undecidability which code might be correct, the superordinate topography code is chosen, e. g. ICD-O-3 C71.9 Brain, NOS. In case of missing topography information, that topography code is assigned which can be derived from the ICD-10 diagnosis (mostly a Cxx.9). In case of two differing diagnoses, the best compatible with the histology code ICD-10 code is chosen. If a diagnosis is not decidable because both could be valid, the umbrella ICD-10 code is assigned, e. g. z. B. D33.9, D43.9 or C71.9. In case of a missing diagnosis the respective ICD-10 code is generated from the ICD-O-3-morphology code and the ICD-O-3-topography code. If ICD-O-3 morphology code and diagnosis code are incompatible, the morphology code overrides the ICD-10 code. If ICD-O-3 topography and diagnosis are incompatible and retrieval of further information is impossible the superordinate ICD-10 code is assigned, e. g. ICD-10 D33.9, D43.9 or C71.9, i. e. unspecified diagnoses. Results/Conclusions: If data are missing, incomplete and/or incompatible in CNS tumour data sets a relatively small number of rules are needed to generate a more complete and qualitively improved data set. Application of these rules facilitates the preparation of a CNS tumour data pool in one’s own cancer registry and for comparison between regional registries. The suggested approach can support clinical and epidemiological research by improving the quality and completeness of cancer registry data. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

173


CONTENTS AUTHOR INDEX

Wissenschaftliches Symposium Immunphänotypisierung

Plenarsitzung Best Abstract

V571

V579

Significance of flow cytomeric MRD quantification in CLL and multiple myeloma

Randomized, open-label, multicenter, phase 3 study of eribulin versus dacarbazine in patients (pts) with leiomyosarcoma (LMS) and adipocytic sarcoma (ADI)

Böttcher S.1 Universitätsklinikum Schleswig-Holstein, Campus Kiel, II. Medizinische Klinik, Kiel, Germany 1

The very sensitive quantification of residual cells after and during treatment is gaining importance in virtually all hematological malignancies as the number of available treatment alternatives and their efficacy increases. While clinical complete remissions (CR) have become attainable in many leukemias and lymphomas, it has become increasingly clear that relapses, likely originating from very few residual neoplastic cells, often occur even in CR patients. Molecular and flow cytometric technologies for sensitive MRD determination have been shown to be of prognostic significance in several incurable mature B-cell malignancies over the last two decades. Amongst those diseases, flow cytometric MRD detection (as opposed to PCR-based approaches) have been applied most broadly in chronic lymphocytic leukemia (CLL) and in multiple myeloma (MM). MRD detection by flow cytometry has benefited from early pivotal studies identifying reliable markers for the identification of the target cell population and for the distinction of malignant cells from their benign counterparts (CLL: CD19, CD5, CD20, CD38, CD81, CD22, CD43, CD79b; MM: CD138, CD38, CD45, CD56, CD19, CD27, CD81). As a general concept MRD detection in both diseases relies on the detection of expression differences of these markers for the identification of the neoplastic cells. International guidelines for standardized flow cytometric MRD detection in CLL and MM are available improving comparability of MRD results between different trials. CLL and MM resemble each other biologically as incurable diseases that, however, can be efficaciously treated using chemotherapy and novel agents. Consequently, MRD has been shown to be of prognostic significance primarily for progression free survival in both malignancies. The prognostic significance of MRD in CLL and MM also shows similarities with regards to the importance of MRD levels, of additional (in particular cytogenetic) risk features and of achieving conventional clinical responses. MRD assessments likely qualify as surrogate efficacy end-points in randomized clinical trials comparing different induction therapies, while their application to tailor treatment decisions outside clinical trials still requires additional data to corroborate the utility of such an approach. Disclosure: Sebastian Böttcher: Financing of Scientific Research: Becton Dickinson; Expert Testimony: Becton Dickinson.

Schöffski P.1, Maki R.2, Italiano A.3, Gelderblom H.4, Choy E.5, Grignani G.6, Camargo V.7, Bauer S.8, Rha S.Y.9, Chawla S.10, Blay J.-Y.11, Hohenberger P.12, D’Adamo D.13, Chen R.13, Chmielowski B.14, Le Cesne A.15, George D.16, Patel S.17 University Hospitals Leuven, Department of General Medical Oncology, Leuven, Belgium, 2Mount Sinai Medical Center, Tisch Cancer Institute, New York, United States, 3Institut Bergonié, Department of Oncology, Bordeaux, France, 4Leiden University Medical Center, Department of Medical Oncology, Leiden, Netherlands, 5Massachusetts General Hospital, Department of Medicine, Boston, United States, 6Fondazione del Piemonte per l’Oncologia IRCC, Department of Medical Oncology, Candiolo, Italy, 7Hospital Sírio-Libanês, Department of Medical Oncology, Sau Paulo, Brazil, 8West German Cancer Center, Essen, Germany, 9Severance Hospital, Sinchon-dong, Korea, Republic of, 10 Sarcoma Oncology Center, Santa Monica, United States, 11Université Claude Bernard & Centre Léon Bérard, Lyon, France, 12Mannheim University Medical Center, Division of Surgical Oncology & Thoracic Surgery, Department of Surgery, Mannheim, Germany, 13Eisai Inc, Woodcliff Lake, United States, 14UCLA Jonsson Comprehensive Cancer, Department of Hematology, Los Angeles, United States, 15Gustave Roussy, Villejuif, France, 16Sarcoma Center and Ludwig Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, United States, 17University of Texas, MD Anderson Cancer Center, Department of Sarcoma Medical Oncology, Houston, United States 1

Introduction: In a phase 2 study of pts with advanced soft tissue sarcoma, 32% and 47% of pts with LMS and ADI respectively, treated with the microtubule dynamics inhibitor eribulin achieved progression-free survival (PFS) at the 12 wk timepoint (Schöffski et al. Lancet Oncol. 2011; NCT00413192). Based on these findings, this phase 3 study (NCT01327885) compared overall survival (OS) in pts with advanced LMS and ADI treated with eribulin or dacarbazine. Methods: Pts aged ≥18 yrs with advanced high/intermediate grade LMS or dedifferentiated, myxoid, round cell or pleomorphic variants of ADI incurable by surgery and/or radiotherapy were enrolled. Pts had ECOG status ≤2 and had received ≥2 standard systemic treatment regimens including an anthracycline. Pts were randomized 1:1 to eribulin (1.4 mg/ m2, IV on D1 and D8) or dacarbazine (850–1200 mg/m2, IV on D1) every 21 days until disease progression. Primary endpoint was OS. Secondary endpoints included PFS, PFS rate at Wk 12 and safety. Results: Overall, 452 pts (67% female; 79% < 65 yrs) were randomized (228 eribulin; 224 dacarbazine). Median OS for eribulin and dacarbazine was 13.5 and 11.5 months, respectively (HR = 0.768, 95% CI 0.618–0.954; P = 0.017). PFS was 2.6 months in both arms (HR = 0.877, 95% CI 0.710– 1.085; P = 0.229). PFS rate at Wk 12 was 33% and 29% for eribulin and dacarbazine, respectively. In eribulin and dacarbazine arms, respectively, 26% and 14% of pts required dose reductions and 8% and 5% discontinued due to treatment-emergent adverse events (TEAEs). TEAEs were more frequent in eribulin than dacarbazine arm, including neutropenia (44% vs 24%), pyrexia (28% vs 14%), peripheral sensory neuropathy (20% vs 4%) and alopecia (35% vs. 3%); as were TEAEs of grade 3 (63% vs 53%), grade 4 (26% vs 20%), and fatal TEAEs (4% vs 1%). Thrombocytopenia was more frequent in dacarbazine than eribulin arm (28% vs 6%). Conclusions: This phase 3 trial of eribulin trial met its primary objective of OS benefit in pretreated pts with advanced LMS or ADI. Eribulin had a toxicity profile consistent with prior experience, with no unexpected or new safety findings. Disclosure: Patrick Schöffski: Financing of Scientific Research: GSK, Novartis, Threshold; Expert Testimony: GSK, Novartis, Bayer, Genetech, Exelixis Shreyakumar Patel: Advisory Role: Advisory Role/Consultant; Expert Testimony: Clinical Trial Funding.

174

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V580

V581

Impact of antibiotic treatment modulating the intestinal microbiota on the efficacy of antineoplastic treatment for chronic lymphatic leukaemia or relapsed lymphoma

KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance

Pflug N.1, Kluth S.1, Vehreschild J.J.1,2, Bahlo J.1, Tacke D.1,2, Biehl L.1,2, Eichhorst B.1, Fischer K.1, Cramer P.1, Fink A.-M.1, von Bergwelt-Baildon M.1, Hallek M.1, Cornely O.A.1,2,3, Vehreschild M.J.G.T.1,2 University Hospital of Cologne, 1st Department of Internal Medicine and Center for Integrated Oncology CIO Köln/Bonn, Cologne, Germany, 2German Centre for Infection Research (DZIF), Site Bonn/Cologne, Cologne, Germany, 3 University of Cologne, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), and Clinical Trials Center Cologne, ZKS Köln, BMBF 01KN1106, Cologne, Germany 1

Introduction: Recent publications have shown reduced anticancer efficacy of cyclophosphamide (CYC) and platinum salts in germ-free mice and animals treated with antibiotics with activity in the Gram-positive spectrum. In recently published mouse models , translocation of Gram-positive commensal bacteria during chemotherapy-associated mucositis and subsequent reprogramming of intratumoral myeloid cells was proposed as the underlying mode of action. To assess this hypothesis in a clinical setting, we identified patients receiving CYC for chronic lymphatic leukaemia (CLL) and receiving cisplatin for relapsed lymphoma. Methods: Data was selected from the CLL8 trial (NCT00281918) and the Cologne Cohort of Neutropenic Patients (CoCoNut, NCT01821456). Relevant antibiotics were defined as substances with primary activity against Gram-positive bacteria. The impact of these antibiotics on response and progression-free survival (PFS) was analysed by Kaplan-Meier methodology and Cox proportional hazards regression analysis. Results: Out of 809 CLL patients analysed, those receiving anti-Gram-positive antibiotics (N = 46/763) were significantly less likely to achieve an overall response (OR 74.3% vs. 90.2%, p = 0.003) or complete response (CR 22.9% vs. 33.8%, p = 0.01). Patients who received anti-Gram-positive antibiotics progressed significantly earlier than did other patients (median PFS 14.1 vs. 44.1 months, p < 0.001). In the regression analysis, administration of anti-Gram-positive antibiotic treatment was identified as an independent prognostic factor for reduced PFS (Hazard ratio (HR) 2.077, p = 0.002). Of 123 patients with relapsed lymphoma those receiving anti-Gram-positive antibiotics (N = 22/101) were significantly less likely to achieve OR (70.3% vs. 45.5%, p = 0.026), and had numerically lower CR rates (15.8% vs. 13.6%, p = 0.076). Patients with anti-Gram-positive antibiotics progressed significantly earlier than those without (median PFS 6.0 vs. 96.0 months, p < 0.001). In the regression analysis, Ann Arbor Stage ≥3 at diagnosis of relapse (HR 2.249, p = 0.011) and use of anti-Gram-positive antibiotics independently predicted reduced PFS (HR 1.876, p = 0.046). Conclusion: Our data supports the negative impact of anti-Gram-positive antibiotics on the anticancer activity of CYC and cisplatin in a clinical setting, and may help design further studies in this area. Disclosure: No conflict of interest disclosed.

Metzgeroth G.1, Schwaab J.1, Umbach R.1, Naumann N.1, Jawhar M.1, Sotlar K.2, Horny H.-P.2, Gaiser T.3, Hofmann W.-K.1, Schnittger S.4, Cross N.C.P.5,6, Fabarius A.1, Reiter A.1 III. Medizinische Klinik, Universitätsmedizin Mannheim der Universität Heidelberg, Mannheim, Germany, 2Pathologisches Institut, Ludwig-MaximiliansUniversität, München, Germany, 3Pathologisches Institut, Universitätsmedizin Mannheim der Universität Heidelberg, Mannheim, Germany, 4MLL Münchner Leukämielabor, München, Germany, 5Wessex Regional Genetics Laboratory, University of Southampton, Salisbury District Hospital, Salisbury, United Kingdom, 6Faculty of Medicine, University of Southampton, Southampton, United Kingdom 1

Introduction: In hypereosinophilia of unknown significance (HEUS), clonality of eosinophils can be proven by identification of acquired chromosome or molecular abnormalities of which the most common is the FIP1L1-PDGFRA (FP) fusion gene. However, a large proportion of cases with clinical signs of an eosinophilia-associated myeloproliferative neoplasm (MPN-eo) are FP negative and show a normal karyotype. Recurrent molecular markers of related MPN subtypes include KIT D816V (systemic mastocytosis, SM) and JAK2 V617F (classical MPN). Methods: We investigated 426 samples (BM/PB) from patients with HEUS for the fusion gene and KIT D816V (quantitative allele specific PCR) or JAK2 V617F mutations and compared clinical and molecular characteristics. Results: Overall, 86 (20%) cases with HEUS were positive for FP (n = 55, 13%), KIT D816V (n = 14, 3%) or JAK2 V617F (n = 17, 4%). While FP and KIT D816V or JAK2 V617F were mutually exclusive, 3 KIT D816V cases were also JAK2 V617F positive. All KIT D816V positive cases were subsequently diagnosed as SM and were therefore analyzed along with 31 additional, well-characterized KIT D816V positive SM patients with eosinophilia (SM-eo) enrolled within the “German Registry on Disorders of Eosinophils and Mast cells”. FP positivity was strongly associated with younger age at diagnosis, male predominance and higher numbers of eosinophils while abdominal lymphadenopathy, ascites and serum tryptase levels >100 µg/l were characteristic for SM-eo. No differences were found regarding white blood cell count, hemoglobin level and splenomegaly. A median of 3 additional mutations, most frequently TET2 and SRSF2, were identified in 12/13 (92%) SM-eo patients indicating a more complex molecular pathogenesis. Median survival was not reached for FP cases but was only 26 and 41 months for KIT D816V positive SM-eo and JAK2 V617F positive MPN-eo, respectively. In KIT D816V and JAK2 V617F positive patients, eosinophilia of ≥2×109/l was identified as discriminator for inferior survival (median 20 months vs. not reached, p = 0.002). Conclusion: We here report on the largest cohort of HEUS patients to be systematically screened for FP, KIT D816V and JAK2 V617F. Despite the rarity of those molecular aberrations, their identification is of clear diagnostic, prognostic and therapeutic relevance because of the potential for targeted therapy. Eosinophilia is significantly associated with poorer outcome in KIT D816V positive SM and JAK2 V617F positive MPN-eo. Disclosure: Georgia Metzgeroth: No conflict of interest disclosed. Andreas Reiter: Advisory Role: Novartis; Financing of Scientific Research: Novartis. V582

The zebrafish homologue of the murine Evi1 gene critically regulates HSC development via the NOTCH pathway Konantz M.1, Müller J.1, Lenard A.1, Alghisi E.1, Esain V.2, Carroll K.J.2, North T.E.2, Lengerke C.1,3 University Hospital Basel, Department of Biomedicine, Basel, Switzerland, Harvard Medical School, BIDMC, Department of Pathology, Boston, United States, 3University Hospital Basel, Clinic for Hematology, Basel, Switzerland 1 2

Introduction: EVI1 is one of the most potent oncogenes associated with myeloid malignancies and is expressed in healthy adult hematopoietic

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

175


CONTENTS AUTHOR INDEX

stem cells (HSCs). Here, we employ the zebrafish (ZF) model to explore the role of ZF evi1 during blood development. Methods: Genetic modifications were obtained by injection of inhibiting morpholino oligonucleotides or mRNA into the ZF zygote or via heatshock inducible transgenic lines. Haematopoiesis was analysed by in situ hybridisation, live cell imaging and flow cytometry on single-cell dissociated transgenic embryos. Sorted cells were further subjected to qRT-PCR analysis. Results: Knockdown of evi1 strongly impaired embryonic myelopoiesis, while not altering primitive erythropoiesis. Furthermore, evi1 morphants showed reduced runx1/cmyb+ HSCs in the AGM, and later on showed less circulating globin+ erythrocytes or lyz+ myeloid cells and impaired megakaryopoiesis. Consistently, lack of ikaros+ lymphocyte precursors and rag1+ T-lymphocytes was observed. Live imaging of double transgenic Tg(Kdrl:mKate-CAAX;cmyb:GFP) embryos indicated reduced emergence of mKate+GFP+ putative HSCs from the ventral wall of the dorsal aorta (DA), suggesting defects in HSC budding from the haemogenic endothelium (HE). Consistently, the DA showed reduced expression of efnb2a and dlc. Furthermore, mKate+GFP+ cells sorted from evi1 morphants showed diminished expression of hematopoietic genes (runx1, cmyb) and enhanced expression of endothelial specific genes (kdrl, flt1, dab) as compared to corresponding control cells, suggesting that evi1 is required for the full transition of HE cells to hematopoietic fate. On the molecular level, evi1 morphants showed reduced levels of notch pathway members, and HSC formation was rescued by conditional heat-shock induction of NICD or its upstream regulator VEGF. Enforced tissue-specific NICD expression in double-transgenic cdh5:Gal4;UAS:NICD fish revealed that spatial expression of NICD is sufficient to rescue the HSC phenotype. Furthermore, the notch target gene gata2 also provided a partial HSC rescue. Conclusion: Taken together, our data suggest that evi1 regulates HSC specification from the HE via activation of NOTCH and downstream gata2. Currently, we analyse in more detail how evi1 regulates NOTCH on the molecular level and use the CRISPR/Cas9 system to generate evi1 mutant fish for further validation of our findings. Disclosure: No conflict of interest disclosed.

ing (NGS). The role of distinct mutational patterns for treatment outcome remains largely unknown. We here analyzed mutational patterns of HPV+ and HPV- tumors and compared results with outcome after uniform chemoradiation. Methods: Archival tumor specimens from 208 patients with LASCCHN of the hypopharynx, oropharynx or oral cavity, all uniformly treated with surgery and adjuvant cisplatin-based radiochemotherapy at one of the eight partner sites of the German Cancer Consortium were included in this study. An in-house gene panel for semiconductor-based sequencing, covering 211 exons from 45 genes frequently altered in SCCHN was used for mutational analysis. Genetic alterations were correlated with HPV status, clinical risk parameters and patient outcome. Findings: Mutational profiles were successfully established for 185 SCCHN cases. Interestingly, HPV+ carcinomas were significantly enriched for activating mutations in driver genes (PIK3CA 27%, KRAS 8%, NRAS 4%, HRAS 2%) compared to HPV- cases (P = 0.002). Conversely, HPV- tumors showed an increased frequency of loss-of-function alterations in tumor suppressor genes (TP53 67%, CDKN2A 30%, PTEN 4%, SMAD4 3%) compared to HPV+ cases (P< 0.001). After a median follow-up of 55 months, detection of alterations in tumor suppressor genes significantly increased the risk of death (HR 2.9, 95% CI 1.5–5.8, P = 0.001), locoregional recurrence (HR 5.4, 95% CI 1.6–18.1, P = 0.006) and distant metastasis (HR 2.3, 95% CI 1.0–5.1, P = 0.04). The occurrence of activating driver gene mutations did not significantly influence outcome in the total cohort of patients, however, they were associated per trend with increased risks of locoregional recurrence and death (HR 3.7, 95% CI 0.7–20.6, P = 0.12) in the subgroup of HPV+p16+ oropharyngeal carcinomas. Conclusion: Activating mutations in driver genes occuring in one third of HPV-driven SCCHN seem to negatively interfere with efficacy of adjuvant cisplatin-based chemoradiation. These genes or their associated signaling pathways might represent therapeutic targets for improving cure rates of HPV+ disease. Disclosure: No conflict of interest disclosed.

V583

Next-generation sequencing of locally advanced squamous cell carcinomas of the head and neck (LASCCHN) for assessment of mutational patterns and their interference with outcome after adjuvant chemoradiation: Results from a multicenter biomarker study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG) Tinhofer I.1,2, Budach V.1,2, Linge A.3, Lohaus F.3, Gkika E.4, Stuschke M.4, Balermpas P.5, Rödel C.5, Avlar M.6, Grosu A.L.6, Abdollahi A.7, Debus J.7, Belka C.8, Pigorsch S.8, Combs S.E.8, Mönnich D.9, Zips D.9, Weichert W.7, Krause M.3, Baumann M.3, DKTK-ROG Charité – Universitätsmedizin Berlin, Klinik für Radioonkologie und Strahlentherapie, Berlin, Germany, 2Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Berlin, Berlin, Germany, 3Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Dresden, Dresden, Germany, 4Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Essen, Essen, Germany, 5Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Frankfurt, Frankfurt, Germany, 6Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Freiburg, Freiburg, Germany, 7Deutsches Krebsforschungszentrum (DKFZ) und Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany, 8Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort München, München, Germany, 9Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort Tübingen, Tübingen, Germany 1

Background: The genetic landscape of SCCHN is currently being unravelled by an increasing number of studies using next-generation sequenc-

176

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V584

Improved outcome with addition of thiotepa and rituximab to methotrexate and cytarabine in primary cns lymphoma: Results of the first randomization of the IELSG-32 trial Illerhaus G.1, Cwynarski K.2, Pulczynski E.3, Ponzoni M.4, Deckert M.5, Politi L.S.4, Fox C.P.6, La Rosé P.7, Ambrosetti A.8, Röth A.9, Hemmaway A.10, Ilariucci F.11, Linton K.M.12, Soffietti R.13, Pukrop T.14, Binder M.15, Balzarotti M.16, Fabbri A.17, Johnson P.18, Gorlov J.S.19, Cavalli F.20, Finke J.21, Schorb E.21, Reni M.22, Zucca E.23, Ferreri A.J.M.24 Klinikum Stuttgart, Hämatologie, Onkologie und Palliativmedizin; Stuttgart Cancer Center, Stuttgart, Germany, 2Royal Free Hospital and UCLH, London, United Kingdom, 3Aarhus University Hospital, Aarhus, Denmark, 4Ospedale San Raffaele Scientific Institute, Milano, Italy, 5Uniklinik Köln, Institut für Neuropathologie, Köln, Germany, 6Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom, 7Universitätsklinikum Jena, Abt. Hämatologie u. internistische Onkologie, Jena, Germany, 8Policlinico G.B. Rossi, Verona, Italy, 9 Universitätsklinikum, Essen, Germany, 10Queen’s Hospital, Romford, United Kingdom, 11Arcispedale Santa Maria Nuova, Reggio Emilia, Italy, 12Christie Hospital NHS Trust, Manchester, United Kingdom, 13AOU Città della salute e della scienza di Torino, Ematologia 2, Torino, Italy, 14Universitätsklinikum Regensburg, Klinik und Poliklinik für Innere Medizin III, Regensburg, Germany, 15 Universitätsklinikum Hamburg Eppendorf, Klinik und Poliklinik für Onkologie, Hämatologie und KMT mit Sektion Pneumologie, Hamburg, Germany, 16Istituto Clinico Humanitas, Milano Rozzano, Italy, 17Azienda Ospedaliera Università Senese, Siena, Italy, 18Southampton General Hospital, Southampton, United Kingdom, 19Rigs Hospital, Copenhagen, Denmark, 20Oncology Institute of Southern Switzerland, Bellinzona, Switzerland, 21Uniklinik Freiburg / Innere Medizin / Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 22San Raffaele Scientific Institute, Milano, Italy, 23Ospedale San Giovanni, Bellinzona, Switzerland, 24San Raffaele Scientific Institute, Unit of Lymphoid Malignancies, Milano, Italy 1

Introduction: Efficacy and safety of additional rituximab (R) ± thiotepa (TT) to high dose methotrexate (MTX) and cytarabine (AraC) in patients with untreated primary CNS lymphoma (PCNSL) was evaluated in an international randomized phase II trial (IELSG #32, NCT01011920) (1st randomization). Induction chemotherapy was followed by either high dose chemotherapy supported by autologous stem cell transplantation (HDT-ASCT) or whole brain irradiation (WBRT) (2nd randomization). Results of the first randomization are presented. Methods: Pts 18–70 ys old and ECOG PS ≤3 (PS ≤2 if age 66–70) with new biopsy-proven PCNSL and measurable disease were randomly assigned to receive 4 courses of MTX 3.5 g/m2 d1 + AraC 2 g/m2 x2/d d2–3 (arm A); or MTX-AraC + R 375 mg/m2 d-5 & 0 (arm B); or MTXAraC-R + TT 30 mg/m2 d4 (arm C). Stem-cells were collected after the 2nd course. Response was assessed after 2nd and 4th courses; pts with responsive disease were further randomized between WBRT and BCNU-TT conditioned ASCT. Histology and neuroimaging were centrally reviewed. Primary endpoints were CRR (1st random) and 2-year FFS (2nd random). Results: 227 pts (median age 58 ys; 18–70) were enrolled in 52 centers in 5 countries; 8 pts were excluded due to misdiagnosis, systemic disease or concomitant cancer. No differences in clinical presentation among the 3 arms (A 75; B 69; C 75) were observed. 733/876 (84%) planned courses were delivered. G4 hematological toxicity was more common in arm C, infective complications were similar in all 3 arms. G4 non-hematological toxicities were rare. Chemotherapy was interrupted due to toxicity in 21 (9%) pts; 13 (6%) pts died of toxicity. ASC were collected in 152/161 (94%) pts. Arm C was significantly more active, with a CRR of 49% and an ORR of 87%; 118 pts (A 35; B 35; C 48) were referred to 2nd random (59 pts/arm). At a median follow-up of 20 months (7–60), 111 pts remain failure-free (A 25; B 37; C 49), with 2-yr FFS of 34 ± 6%, 52 ± 6% and 64 ± 6% (p = 0.0006), respectively. 124 pts are alive (A 31; B 41; C 52), with 2-yr OS of 40 ± 6%, 58 ± 6% and 66 ± 6% (p = 0.01), respectively. Conclusions: Apart from greater hematological toxicity, the addition of thiotepa and rituximab to MTX-AraC (=MATRix regimen) was not associated with higher rates of severe complications but resulted in significantly improved response, FFS and OS rates in PCNSL patients. Disclosure: No conflict of interest disclosed.

Abstracts

Wissenschaftliches Symposium

Multiples Myelom Risiko- und altersadaptiertes Vorgehen V585

Therapeutic approaches in multiple myeloma: Precision medicine or one-size fits all? Raab M.S.1,2 Universitaetsklinikum Heidelberg, Medizinische Klinik V, Hämatologie, Onkologie und Rheumatologie, Heidelberg, Germany, 2Deutsches Krebsforschungszentrum, Max-Eder-Einheit G170, Heidelberg, Germany 1

A better understanding of the impact of patient-, disease- and treatment-specific factors on survival and quality-of-life has facilitated the development of a personalised approach to the treatment of myeloma. Age as a surrogate of performance and constitutional reserve has long been used to determine eligibility for transplantation as well as being a component of a risk-versus-benefit-based approach to the intensity of therapy e.g. fit, unfit, and frail. Equally, knowledge of both patient co-morbidities and agent-specific toxicities allows for a more rational choice of therapy. Appropriate caution is required, for example, in the use of anthracyclines or carfilzomib in those with pre-existing cardiac disease or agents known to cause neuropathy in patients with long-standing diabetes. Recent years have also brought greater insight into the long-recognised genetic and clinical heterogeneity of this malignancy. Current guidelines (IMWG, mSMART, etc.) recognise the value of risk-based approaches based on FISH cytogenetic analysis. The differential sensitivity of particular disease subtypes to individual drug classes has also become apparent e.g. the efficacy of bortezomib in t(4;14) myeloma and, conversely, the limited role for conventional chemotherapeutic agents in those with disease harboring a 17p deletion. Immunophenotyping and sequencing-based approaches are close to offering profiles for use in clinical practice, raising the prospect of the ‘real-time’ targeting of uncommon patient-specific molecular markers by direct and indirect methods. While the majority of genetic alterations have recently been found to be limited to relatively small subsets of patients, the overall genome-wide distribution is strikingly clustered on specific signaling cascades and intracellular mechanisms, indicating these mechanisms to be of pathophysiological relevance and therapeutic interest. Potential targets for precision therapy based on molecular profiling include the signaling cascades downstream of the RAS/RAF kinases, the NFkB pathway, protein homeostasis, or therapies based on epigenetic alterations in tumour cells. However, the design of clinical trials will need to evolve in order to keep pace with the greater specificity offered by such personalised therapy. In summary, personalized treatment strategies are already widely applied in multiple myeloma and will become the standard in the near future. Disclosure: Marc Raab: Advisory Role: Amgen, Celgene, BMS, Novartis; Expert Testimony: Novartis, Morphosys. V587

Novel immune-based strategies for multiple myeloma therapy: mAbs and CARs – hope or hype? Hudecek M.1 Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany 1

Adoptive immunotherapy with monoclonal antibodies (mAbs) and T cells that were modified by gene-transfer to express a tumor-targeting synthetic chimeric antigen receptor (CAR) is being investigated as a novel and transformative way for treating multiple myeloma. MAbs targeting the CD38 and CS1 antigens have entered clinical evaluation with encouraging preliminary results. First clinical data on the use of CAR-modified T cells targeting the CD19 and BCMA antigens are also emerging. We will discuss the latest clinical and pre-clinical developments with a focus on rational target antigen selection to maximize chances for inducing durable complete responses without toxicity. An ongoing effort in the field is to

Oncol Res Treat 2015;38(suppl 5):1–270

177


CONTENTS AUTHOR INDEX

identify and validate alternative myeloma antigens that can be targeted without toxicity to essential normal tissues. We will present an update on the pre-clinical pipeline of myeloma-reactive CARs that are under development in our laboratory and in preparation to enter the clinical arena. Disclosure: No conflict of interest disclosed. V588

Treatment oft the frail myeloma patient Weisel K.C.1 Universitätsklinik Tübingen, Hämatologie und Onkologie, Tübingen, Germany

1

Multiple Myeloma (MM) predominantely affects elderly patients, which > 60% of diagnoses and nearly 75% of deaths occuring in those over 65 years of age. Although novel agents have substantially improved MM outcome, patients over 70 years of age benefit less from new treatments. The elderly patient population per se is heterogeneous, consisting of “elderly-fit“ and “elderly-frail“ patients. Frailty is a state of cumulative decline in many physiological systems, resulting in a diminished resistance to stressors, such as cancer and its treatment. Older adults are commonly excluded from cancer clinical trials. Although age cutoffs are being removed from clinical trial designs, occult factors exclude older patients from clinical trial enrollment such as concomitant comorbidities and abnormal laboratory tests. Deciding how aggressively to treat MM in a senior patient is a clinical dilemma that requires a thorough understanding of the indications and possible outcomes for the medical interventions being considered. Recently, Palumbo and coworkers established a geriatic assessment (GA) score for elderly newly diagnosed myeloma patients that combines age, functional status, and comorbidities which can predict survival and toxicity. This frailty score predicts mortality and the risk of toxicity in elderly myeloma patients, which will be implemented in future trial designs. The standard approved treatments for newly diagnosed elderly MM include so far triple combinations with melphalan-prednisone-bortezomib (VMP) or melphalan-prednisone-thalidomide (MPT). So far, no changes in dose and schedule are approved according to age or performance status. Unfortunately, these standard schedules induced a high rate of adverse events with survival benefit inferior in patients >75 years. Recently, continuous treatment with oral Lenalidomide and Dexamethasone (Rd) significantly improved outcome and reduced the toxicities compared with the standard MPT. These data show that doublet regimens can be at least as effective as triplet combinations with a better safety profile. In conclusion, the approach to therapy in older adults with MM must be individualized, based on the patient’s disease characteristics and on the overall health, which may be summarized by using GA. It is needed to develop a personalized approach to therapy to optimize the chance for control of disease while minimizing risk of toxicity and helping the individual meet their goals for their MM treatment. Disclosure: Katja Weisel: Advisory Role: Amgen, Bristol Myers Squibb, Celgene, Janssen, NOXXON, Onyx; Financing of Scientific Research: Amgen, Bristol Myers Squibb, Celgene, Janssen, Onyx; Expert Testimony: Celgene, Janssen; Other Financial Relationships: Amgen, Bristol Myers Squibb, Celgene, Janssen, Novartis, Onyx, Takeda.

Fortbildung

NHL indolent unterschiedliche Krankheiten – Gemeinsamkeiten V589

Challenges in the treatment of follicular lymphomas Ghielmini M.1, SAKK Istituto Oncologico della Svizzera Italiana, Oncologia Medica, Bellinzona, Switzerland 1

Follicular lymphoma is the most frequent lymphoma in the western world after diffuse large B-cell lymphoma. Its median survival after the advent

178

Oncol Res Treat 2015;38(suppl 5):1–270

of new treatment modalities and better supportive care has increased to approximately 14 years. Early stage disease is usually treated with involved field radiotherapy. For advanced stage, a watch and wait policy is often advocated. Studies comparing the response rate at diagnosis or after a period of watch and wait have not shown a difference in response rate. Many treatments were used in first-line for follicular lymphoma, none of which could show to be better than the other. Historically single agent treatments as Chlorambucile or Cyclophosphamide were used, and in the next decades they were combined with Prednisolon, Vincristine, and subsequently also Doxorubicine as in aggressive lymphomas. The experience of these decades showed that the more aggressive the regimen, the longer the disease free survival. Nevertheless, this prolongation of disease free survival could never show to significantly translate in improved overall survival, except maybe for grade 3B FL. Because Bendamustine has substantially less side-effects than CHOP, the combination of Bendamustine and rituximab constitutes today a very good option for patients with follicular lymphoma. Maintenance with one infusion of rituximab every 2–3 months improves EFS but its impact on OS is less well established. Patients in relapse can be treated in many ways, including the repetition of the first line treatment if the first remission was long, or non-cross resistant chemotherapy regimens. New drugs, as PI3K inhibitors have shown excellent results in this indication and were recently approved for this indication. High-dose therapy with autologous peripheral stem cells rescue is frequently used for patients in first or second relapse and, in this setting, could show to improve survival. Allogeneic transplantation associates the debulking effect of high-dose chemotherapy to the immunologic draft versus lymphoma effect. It is therefore capable of obtaining a plateau of the survival curve and is indeed probably the only treatment modality with a chance of cure. Unfortunately, this modality, despite of the better results with the reduced intensity conditioning method is still bound to a high early mortality rate. Disclosure: Michele Ghielmini: Advisory Role: Gilead, Abvie; Financing of Scientific Research: Roche, Cellgene, Janssen, Gilead, Mundipharma; Expert Testimony: Roche. V591

The many faces of marginal-zone lymphoma Raderer M.1 Medizinische Universität Wien, Innere Medizin I, Onkologie, Wien, Austria

1

Marginal zone lymphomas (MZL) according to the current WHO-classification comprise 3 different entities, with two of them being extranodal (i.e. extranodal marginal zone B-cell lymphoma of the mucosa-associated lymphoid tisse – MALT lymphoma and splenic marginal zone lymphomas) and the rare minority being of nodal origin. The latter are mostly diagnosed by excluding an extranodal origin of this CD79a, CD20 positive and CD5-CD10-and CD23- disease composed of small centrocyte-like cells thought to arise from marginal zone cells closely related to the plasma cell. Thus, the phenomenon of plasmacytic differentiation has been described especially in MALT lymphomas, sometimes erroneously leading to diagnosis of extramedullary plasmocytoma. While nodal MZL is thought to be more aggressive than the extranodal MZL variants, little data on therapy as a distinct entity exist, as in clinical practice nodal MZL is being treated according to guidelines estabilshed for follicular lymphoma. In addition, splenic marginal zone lymphoma is also increasingly being treated with regimens including rituximab and bendamustine, slowly replacing the standard approache of splenectomy at least in Southern European countries. Both entities have been linked to infection with Hepatitis C virus, and antiviral therapy may constitute an effective first line therapy resulting in regression of the lymphoma. MALT-lymphoma, however, is characterized more by differences than common features with other indolent lymphomas, at least at initial diagnosis. Especially gastric MALT lymphoma is the prototype of an antigen driven tumor, with Helicobacter pylori (HP) being the most common

Abstracts


CONTENTS AUTHOR INDEX

causative agent. As a consequence, HP-eradication is recommended as first line therapy irrespective of stage in gastric MALT lymphoma (but not extragastric MALT lymphoma), resulting in regression of the lymphoma in up to 75% of patients. The presence of underlying autoimmune diseases as well as the MALT lymphoma specific t(11;18)(q21;q21) translocation have been reported as adverse prognostic factors in MALT lymphoma. However, the rate of HP-negative patients is increasing (25–50% in current series), and response to antibiotics in 15–46% has been reported. Also for ocular adnexal MALT lymphoma, doxycycline is an accepted first line approach. Currently, systemic therapies are increasingly being used also in lolalized MALT lymphoma, and the results and the current state of the art will be summed up. Disclosure: Markus Raderer: Financing of Scientific Research: Roche, Novartis, Ipsen, Celgene, Mundipharma, Bayer, Eisai, Gilead.

Wissenschaftliches Symposium Metastasiertes Mammakarzinom V597

What is the role of resection of primary tumor in metastatic breast cancer? What is the role of cancer stem cells and circulating tumor cells and clinical state of the art Balic M.1 Medizinische Universität, Graz, Austria

1

Based on retrospective analyses of primary tumor surgery in metastatic patients, few prospective clinical trials in primary metastatic breast cancer have been initiated to evaluate the impact of primary tumor surgery on the outcome of patients. First analyses have suggested that a proportion of patients might benefit from such an approach and for at least a proportion of patients surgery of primary tumors may be deteriorating. ABCSG 28 is an ongoing Austrian multicentric prospective study addressing the impact of primary tumor surgery in primary metastatic breast cancer patients. In addition to the clinical question translational research evaluates the role of breast cancer stem cells, circulating tumor cells and circulating cell free DNA to help identify biomarkers associated with progression. Recent findings have revealed cancer as a disease with remarkable heterogeneity and complexity. Specifically, intratumoral heterogeneity has been associated with therapeutic failure and drug resistance and poses considerable clinical challenges to finding effective treatment modalities. Assessing tumor heterogeneity is clinically important for developing novel and efficient targeted therapy concepts. Circulating tumor cells and circulating cell free DNA have emerged as surrogate biomarkers to study metastasis and assess tumor heterogeneity. Clinical studies like ABCSG28 offer an opportunity to evaluate for heterogeneity. Disclosure: No conflict of interest disclosed.

Abstracts

Freier Vortrag CML I V600

Responses to Ponatinib at early landmark time points are associated with Progression-Free Survival (PFS) and Overall Survival (OS) in heavily pretreated Chronic-Phase (CP) Chronic Myeloid Leukemia (CML) patients (Pts) Müller M.C.1, Baccarani M.2, Deininger M.W.3, Guilhot F.4, Hochhaus A.5, Hughes T.P.6, Shah N.P.7, Talpaz M.8, Lustgarten S.9, Rivera V.M.9, Clackson T.9, Haluska F.G.9, Cortes J.E.10 Universitätsmedizin Mannheim, Mannheim, Germany, 2University of Bologna, Bologna, Italy, 3Huntsman Cancer Institute, University of Utah, Salt Lake City, United States, 4Inserm CIC 1402, CHU de Poitiers, Poitiers, France, 5Jena University Hospital, Jena, Germany, 6University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, Australia, 7University of California San Francisco, San Francisco, United States, 8Comprehensive Cancer Center, University of Michigan, Ann Arbor, United States, 9ARIAD Pharmaceuticals, Inc., Cambridge, United States, 10The University of Texas MD Anderson Cancer Center, Houston, United States 1

Introduction: Responses at early landmark time points predict positive long-term outcomes with tyrosine kinase inhibitors (TKIs) in first- and second-line settings. Landmark analyses in pts treated with multiple prior TKIs are limited. Ponatinib, a potent oral TKI, is approved for use in pts with refractory CML or Ph+ acute lymphoblastic leukemia and those with the T315I mutation. Here, the impact of early responses to ponatinib on long-term outcomes in CP-CML pts in the ongoing PACE trial is evaluated. Methods: The retrospective landmark analyses excluded pts with response at baseline, missing or unevaluable assessments at landmark time (< 20 metaphases examined for complete cytogenetic response [CCyR]; < 13 for major cytogenetic response [MCyR]), or progression (for PFS) or discontinuation before landmark time. Cytogenetic responses (MCyR/CCyR, ≤35%/0% Ph+ metaphases) and molecular responses (BCR-ABL ≤0.1% [major molecular response, MMR], ≤1%, and ≤10% on the International Scale) at 3 and 6 months and their association with PFS/OS (Kaplan-Meier estimates) and BCR-ABL ≤0.0032% (MR4.5) are presented. Results: 267 CP-CML pts were evaluated; 60% received ≥3 prior TKIs, median age was 60 (18–94) years, and median time from diagnosis was 7 (0.5–27) years. As of 6 October 2014, median follow-up was 38.4 (0.1– 48.6) months. Responders at 3 months (MCyR, 48%; CCyR, 39%; BCRABL ≤10%, 49%; BCR-ABL ≤1%, 34%; MMR, 14%) typically had better PFS/OS after 2 years than nonresponders, although PFS/OS was high regardless (Fig 1). Molecular response at 3 months predicted MR4.5 over time. Response at 6 months (MCyR, 62%; CCyR, 52%; BCR-ABL ≤10%, 55%; BCR-ABL ≤1%, 49%; MMR, 29%) was also positively associated with PFS/OS after 2 years (Fig 2).

Oncol Res Treat 2015;38(suppl 5):1–270

179


CONTENTS AUTHOR INDEX

Fig. 2.

Fig. 1.

Conclusions: Rapid, deep responses with ponatinib were associated with improved long-term outcomes in this heavily pretreated population. Disclosure: Martin Müller: Financing of Scientific Research: ARIAD, Novartis, BMS; Expert Testimony: ARIAD, Novartis, BMS; Other Financial Relationships: Consultant: ARIAD, Novartis, BMS. Jorge Cortes: Expert Testimony: ARIAD, BMS, Novartis, Pfizer, Teva; Other Financial Relationships: Consultant: ARIAD, BMS, Novartis, Pfizer, Teva.

180

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V601

V602

Impact of unbalanced major and minor route karyotypes at diagnosis on prognosis of CML

Defining a failure time point for major molecular remission in chronic myeloid leukemia: Results of the randomized CML-study IV

Dietz C.T.1, Kalmanti L.1, Fabarius A.1, Lauseker M.2, Haferlach C.3, Schlegelberger B.4, Jotterand M.5, Müller M.C.1, Hänel M.6, Köhne C.-H.7, Goebeler M.-E.8, Pfreundschuh M.9, Balleisen L.10, Burchert A.11, Krause S.W.12, Edinger M.13, Pfirrmann M.2, Hasford J.2, Hochhaus A.14, Saußele S.1, Hehlmann R.1, for the SAKK and the German CML Study Group Medizinische Fakultät Mannheim der Ruprecht-Karls-Universität Heidelberg, III. Medizinische Klinik, Mannheim, Germany, 2Ludwig-MaximiliansUniversität, Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie (IBE), München, Germany, 3MLL Münchner Leukämielabor, München, Germany, 4Medizinische Hochschule, Institut für Humangenetik, Hannover, Germany, 5Centre Hospitalier Universitaire Vaudois (CHUV), Service de génétique médicale, Lausanne, Switzerland, 6Klinikum, Klinik für Innere Medizin III, Chemnitz, Germany, 7Klinikum, Klinik für Onkologie und Hämatologie, Oldenburg, Germany, 8Klinikum, Medizinische Klinik und Poliklinik II, Würzburg, Germany, 9Universitätsklinikum des Saarlandes, Klinik für Innere Medizin I, Homburg, Germany, 10Evangelisches Krankenhaus, Abteilung für Hämatologie-Onkologie, Hamm, Germany, 11Universitätsklinikum Gießen und Marburg, Klinik für Innere Medizin, Schwerpunkt Hämatologie, Onkologie und Immunologie, Marburg, Germany, 12Universitätsklinikum, Medizinische Klinik 5, Erlangen, Germany, 13Universitätsklinikum, Klinik und Poliklinik für Innere Medizin III, Regensburg, Germany, 14Universitätsklinikum, Abteilung für Hämatologie/Onkologie, Jena, Germany 1

Background: Major route additional cytogenetic aberrations (ACA) at diagnosis of chronic myeloid leukemia (CML) indicate an increased risk of progression and shorter survival. Since major route ACA are unbalanced aberrations (except inv(3) and t(3;21) and since patients with minor route ACA may also progress, the question arose whether a more generally defined group of chromosomal abnormalities such as unbalanced aberrations rather than only the specific major route ACA indicate progression. Methods: On the basis of 1348 Philadelphia (Ph) chromosome positive chronic phase patients of the randomized CML-study IV we examined the impact of unbalanced minor route ACA at diagnosis in comparison to major route ACA on prognosis. For cytogenetic analyses of at least 20 Giemsa(G)-banded or Reverse(R)-banded bone marrow metaphases at diagnosis were analyzed. Patients with cytogenetic aberrations in Ph-negative clones at diagnosis were excluded from this analysis. Patients with constitutional changes were assigned to the group with standard translocation t(9;22)(q34;q11) or variant translocation t(v;22). In patients showing a complex aberrant karyotype G- or R-banding analysis was combined with m-FISH analysis. Cytogenetic remission was defined according to the ELN recommendations. Results: At diagnosis, 1175 patients (87%) had a translocation t(9;22) (q34;q11) and 74 (5.5%) a variant translocation t(v;22) only, while a loss of the Y-chromosome (-Y) was present in 44 (3.3%), balanced or unbalanced minor route ACA each in 17 (1.3% each) and major route ACA in 21 (1.6%) patients. Patients with unbalanced minor route ACA achieved complete cytogenetic remission, major molecular remission, progression-free survival (PFS) and overall survival (OS) at similar rates as did patients with t(9;22), t(v;22), -Y, and balanced minor route karyotypes. Only patients with major route ACA had a shorter OS and PFS than all other groups (p < 0.005 for all pairwise comparisons with major route). Five year survival probabilities were for t(9;22): 91.4% (95% CI 89.5–93.1), t(v; 22): 87% (77.2– 94.3), – Y: 89.0% (76.7–97.0), balanced: 100%, unbalanced minor route: 92.3% (72.4–100), major route: 52.2% (28.2–75.5). Conclusions: Only major route, but not unbalanced minor route ACA at diagnosis have a negative impact on prognosis of CML. Disclosure: No conflict of interest disclosed.

Saußele S.1, Hehlmann R.1, Dietz C.T.1, Schnittger S.2, Fabarius A.1, Kohlbrenner K.1, Hanfstein B.1, Proetel U.1, Rinaldetti S.1, Einsele H.3, Falge C.4, Kanz L.5, Neubauer A.6, Kneba M.7, Stegelmann F.8, Pfreundschuh M.9, Waller C.F.10, Baerlocher G.M.11, Heim D.12, Krause S.W.13, Hofmann W.-K.1, Hasford J.14, Pfirrmann M.14, Hochhaus A.15, Müller M.C.1, Lauseker M.14, CML-Studiengruppe und SAKK Medizinische Fakultät Mannheim der Ruprecht-Karls-Universität Heidelberg, III. Medizinische Klinik, Mannheim, Germany, 2MLL Münchner Leukämie Labor, München, Germany, 3Universitätsklinikum, Medizinische Klinik II, Würzburg, Germany, 4Klinikum Nürnberg Nord, Medizinische Klinik 5, Nürnberg, Germany, 5Universitätsklinikum, Innere Medizin II, Tübingen, Germany, 6 Universitätsklinikum Gießen und Marburg, Klinik für Hämatologie, Zentrum Innere Medizin, Marburg, Germany, 7Universitätsklinikum Schleswig-Holstein, II. Medizinische Klinik und Poliklinik, Kiel, Germany, 8Universitätsklinikum, Klinik für Innere Medizin III, Ulm, Germany, 9Universitätsklinikum des Saarlandes, Klinik für Innere Medizin I, Homburg, Germany, 10Universitätsklinikum, Klinik für Innere Medizin I, Freiburg, Germany, 11Inselspital, Universitätsklinik für Hämatologie und hämatologisches Zentrallabor, Bern, Switzerland, 12 Universitätsspital, Klinik für Hämatologie, Basel, Switzerland, 13 Universitätsklinikum, Medizinische Klinik 5, Erlangen, Germany, 14LudwigMaximilians-Universität, Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie (IBE), München, Germany, 15Universitätsklinikum, Klinik für Innere Medizin II, Abt. Hämatologie und Intern. Onkologie, Jena, Germany 1

Introduction: In the current ELN recommendations the optimal time point to achieve major molecular remission (MMR) is defined at 12 months after diagnosis of chronic myeloid leukemia (CML). Lack of MMR is not a failure criterion at any time point leading to uncertainties when to change therapy in CML patients not reaching MMR after 12 months. We sought to evaluate a failure time point (FTP) for MMR using data of the CML-Study IV, a randomized five-arm trial designed to optimize imatinib therapy. Methods: Patients with valid molecular analysis and on imatinib-based treatment were divided randomly into a learning (LS) and a validation sample (VS). Between 0.5 and 5 years after diagnosis, monthly landmarks were defined for the molecular remission levels of the LS. The best prediction time was found via landmarking in combination with a bootstrap approach. For the FTP analysis, a Cox model was used to define the time to progression. The hazard ratios (HR) of each landmark were treated as dependent on the HRs of the other landmarks, using a cubic smoothing function. The minimum of this function was considered to be the best predictor of progression-free survival (PFS). For this time point, landmark analysis of PFS was performed in the VS. For the evaluation of the optimal time point of achieving a MMR the same analysis was done to define the time to MR4.5 or deeper. Results: 1228 out of 1551 patients were randomly allocated to the LS (n = 819) and to the VS (n = 409). The minimum of the cubic function of the HRs was found for MMR at 2.67 years with a HR of 0.24 (compared to patients without any remission) and 0.53 compared to those in MR2. The validation was done with 2.75 instead of 2.67 years since the time interval for molecular evaluation in the study is 3 months. 348 of the 409 patients of the VS were still at risk at the FTP and evaluable. A significant PFS advantage for patients in MMR could be demonstrated (p = 0.022). For the optimal MMR analysis no singular time point could be calculated as the earlier a MMR was reached the higher was the chance to achieve a MR4.5. Conclusions: In this model, an FTP to predict PFS in patients with MMR was defined at 2.75 years after diagnosis and could be validated as significant. Nevertheless, patients being in MMR had a lower risk of progression than patients not being in MMR on any other time point as well. With this model we can give hints when to define MMR as failure and a change in therapy should be considered. Disclosure: Susanne Saußele: Financing of Scientific Research: Novartis, BMS, Pfizer, Ariad; Expert Testimony: Novartis, BMS. Michael Lauseker: No conflict of interest disclosed.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

181


CONTENTS AUTHOR INDEX

V603

V604

Validation of a new prognostic score for survival discriminating three groups with different probabilities of dying of chronic myeloid leukaemia in patients with first-line imatinib treatment

Ponatinib efficacy and safety in heavily pretreated leukemia patients (Pts) in the PACE trial at 3 years

Pfirrmann M.1, Hasford J.1, Turkina A.2, Prejzner W.3, Zackova D.4, Colita A.5, Zaritskey A.6, Steegmann J.-L.7, Indrak K.8, Hehlmann R.9, Baccarani M.10, for the EUTOS Investigators Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie (IBE), Ludwig-Maximilians-Universität, München, Germany, 2 National Research Center for Hematology, Moscow, Russian Federation, 3 Department of Hematology, Medical University of Gdansk, Gdansk, Poland, 4Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Masaryk University, Brno, Czech Republic, 5 Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania, 6St Petersburg State Medical University, St Petersburg, Russian Federation, 7Department of Hematology, Hospital Universitario de la Princesa, Madrid, Spain, 8Department of Hemato-oncology, University Palackianae, Olomouc, Czech Republic, 9III. Medizinische Klinik, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany, 10Department of Hematology and Oncology L. and A. Seragnoli, S Orsola-Malpighi Hospital, Bologna, Italy 1

Introduction: For the identification of 3 risk groups with clinically relevant differences in the cumulative incidence probabilities (CIPs) of dying of chronic myeloid leukaemia (CML), a new survival score was developed in the in-study section of the European Treatment and Outcome Study (EUTOS) registry. The score consists of the factors age, spleen size, platelet count and blasts in peripheral blood. In the out-study section of the registry, where patients were also prospectively registered but not part of clinical trials, we aimed at validation of the new EUTOS survival score and its comparison with the Sokal, the Euro, and the EUTOS score. Methods: Survival was measured from the start of imatinib treatment and censored at the time of allogeneic stem cell transplantation (SCT) in first CP. Only death after recorded disease progression was regarded as CML-specific death. CIPs of dying of CML were estimated, treating death due to any other cause as a competing event. All prognostic scores were calculated at diagnosis of CML. CIPs of different risk groups were compared with the Gray test. Level of significance was 0.05. Results: In 1,120 out-study patients, all prognostic scores were evaluable. Median observation time was 5.6 years. Without prior SCT in first CP, 116 patients died. In 56 cases (48% of 116), cause of death was CML. Six-year CIP of death due to CML was 5% [95%-confidence interval (CI): 4–7%]. The EUTOS survival score identified 681 low-risk (61% of 1,120), 302 intermediate- (27%), and 137 high-risk patients (12%). Their 6-year CIPs of dying of CML were 3% [95%-CI: 2–5%], 6% [95%-CI: 3–9%], and 17% [95%-CI: 11–25%]. It was the only score to identify three risk groups with pairwise significantly different CIPs of dying of CML. Compared with the Sokal and the Euro score, the new score identified a much larger group of low-risk patients and had the highest subdistribution hazard ratio of the high- to the low-risk group [6.746; 95%-CI: 3.614–12.592] and thus, the largest prognostic difference between these groups of all 4 scores. Conclusions: The new EUTOS survival score showed the best prognostic discrimination also in the out-study patients. Validation in the independent out-study data was successful. In this low-risk group, the new score showed an excellent long-term outcome when starting treatment with imatinib. Worse results for the high-risk patients make an upfront comparison between different tyrosine kinase inhibitors desirable. Disclosure: Markus Pfirrmann: Expert Testimony: Das EUTOS-Projekt wurde durch Novartis Oncology Europe unterstützt. Michele Baccarani: Expert Testimony: Das EUTOS-Projekt wurde durch Novartis Oncology Europe unterstützt.

182

Oncol Res Treat 2015;38(suppl 5):1–270

le Coutre P.D.1, Cortes J.E.2, Kim D.-W.3, Pinilla-Ibarz J.4, Paquette R.5, Chuah C.6, Nicolini F.E.7, Apperley J.F.8, Khoury H.J.9, Talpaz M.10, DeAngelo D.J.11, Abruzzese E.12, Rea D.13, Baccarani M.14, Müller M.C.15, Gambacorti-Passerini C.16, Lustgarten S.17, Rivera V.M.17, Clackson T.17, Haluska F.G.17, Guilhot F.18, Deininger M.W.19, Hochhaus A.20, Hughes T.P.21, Shah N.P.22, Kantarjian H.M.2 Charité Universitätsmedizin, Berlin, Germany, 2The University of Texas MD Anderson Cancer Center, Houston, United States, 3Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea, Republic of, 4H. Lee Moffitt Cancer Center & Research Institute, Tampa, United States, 5Ronald Reagan UCLA Medical Center, University of California, Los Angeles, United States, 6Singapore General Hospital and Duke-National University of Singapore Graduate Medical School, Singapore, Singapore, 7Centre Hospitalier Lyon Sud, Pierre Bénite, Lyon, France, 8Centre for Haematology, Imperial College, London, United Kingdom, 9Winship Cancer Institute of Emory University, Atlanta, United States, 10 Comprehensive Cancer Center, University of Michigan, Ann Arbor, United States, 11Dana-Farber Cancer Institute, Boston, United States, 12S. Eugenio Hospital, Tor Vergata University, Rome, Italy, 13Hôpital Saint-Louis, Paris, France, 14 University of Bologna, Bologna, Italy, 15Universitätsmedizin, Mannheim, Germany, 16Azienda Ospedaliera San Gerardo/University of Milano-Bicocca, Monza, Italy, 17ARIAD Pharmaceuticals, Inc., Cambridge, United States, 18Inserm CIC 1402, CHU de Poitiers, Poitiers, France, 19Huntsman Cancer Institute, University of Utah, Salt Lake City, United States, 20Jena University Hospital, Jena, Germany, 21University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, Australia, 22University of California San Francisco, San Francisco, United States 1

Introduction: Ponatinib, an approved oral tyrosine kinase inhibitor (TKI), has potent activity against native BCR-ABL and resistant mutants, including the T315I mutant. Methods: The phase 2 PACE trial (NCT01207440) evaluated efficacy and safety of ponatinib (starting dose 45 mg qd) in pts with CML or Ph+ ALL resistant/intolerant to dasatinib/nilotinib or with T315I. Of 449 treated pts, 58% received ≥3 prior TKIs. Arterial occlusive events (AOEs) led to dose reduction recommendations in October 2013. Exposure-adjusted incidence rates of new AOEs were calculated as: (number of first events in interval)/(total exposure for interval in pt-years) × 100. Pts with prior events were excluded from later intervals. Results: As of 6 October 2014, 33% of all pts and 45% of CP-CML pts remained on study. Median follow-up was 34.2 (0.1–48.6) months for all pts and 38.4 (0.1–48.6) months for CP-CML pts. The main reasons for discontinuation were progression (21% overall, 9% CP-CML) and adverse events (AEs) (15% overall, 17% CP-CML). For CP-CML, response rates were: MCyR, 59%; MMR or better, 39%; MR4.5, 22%. At 3 years an estimated 83% of responders remained in MCyR, and estimated PFS/OS rates were 61%/82%. For AP-CML, BP-CML, and Ph+ ALL, estimated OS rates at 3 years were 59%, 9%, and 16%, respectively. Treatment-emergent AEs in ≥25% of pts were: thrombocytopenia, 43%; abdominal pain, 42%; rash, 41%; constipation, 37%; headache, 37%; dry skin, 36%; fatigue, 30%; pyrexia, 29%; arthralgia, 29%; hypertension, 28%; nausea, 28%; neutropenia, 25%. AOE rates were 22%/17% (AEs/serious AEs), including cardiovascular (12%/8%), cerebrovascular (8%/6%), and peripheral vascular (8%/6%) events; venous thromboembolic event (VTE) rates were 5%/4%. Of the 99 pts with AOEs, 42 remained on study and 33 maintained MCyR. Exposure-adjusted incidence rates of new AOEs/VTEs (events per 100 ptyears) were 14.5/3.5 in Year 1, 14.1/1.8 in Year 2, and 10.8/1.8 in Year 3. A year after recommended dose reduction, 61/64 (95%) CP-CML pts maintained MCyR, 44/47 (94%) CP-CML pts maintained MMR, and 5/70 (7%) ongoing pts without a prior AOE had an AOE. Conclusions: In PACE, ponatinib continues to provide deep, durable responses in heavily pretreated pts, particularly those with CP-CML. A year after recommended dose reduction, responses were maintained and incidence of new AOEs was lower than in previous years. A dose-ranging trial to evaluate benefits and risks of ponatinib in refractory CML will open soon.

Abstracts


CONTENTS AUTHOR INDEX

Disclosure: Philipp le Coutre: Financing of Scientific Research: ARIAD Pharmaceuticals, Inc., Novartis, BMS, Pfizer. Hagop Kantarjian: Expert Testimony: ARIAD Pharmaceuticals, Inc., Pfizer, Amgen.

tigue and biochemical AEs and can be addressed by prolongation of the treatment interval with PEG-IFNα2b.

V605

Freier Vortrag

Feasibility of first line nilotinib plus Peginterferon-α2b in chronic phase of chronic myeloid leukemia (CML): Results of the pilot phase of the German CML V (TIGER)-study Schenk T.1, Eigendorff E.1, Brümmendorf T.H.2, Burchert A.3, le Coutre P.4, Saußele S.5, Stegelmann F.6, Hänel M.7, Staib P.8, Lindemann H.-W.9, Lange E.10, Müller L.11, Schwarzer A.12, Jentsch-Ullrich K.13, Schenk M.14, Greiner J.15, Fabisch C.1, Pfirrmann M.16, Hochhaus A.1, for the Geman CML Study Group Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany, 2Universitätsklinikum RWTH Aachen, Abteilung für Onkologie, Hämatologie und SZT, Aachen, Germany, 3 Universitätsklinikum Gießen und Marburg, Klinik für Hämatologie, Onkologie und Immunologie, Marburg, Germany, 4Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany, 5Universitätsmedizin, Mannheim, Germany, 6Klinik für Innere Medizin III, Universitätsklinikum, Ulm, Germany, 7Klinik für Innere Medizin III, Klinikum Chemnitz gGmbH, Chemnitz, Germany, 8St.-Antonius-Hospital, Klinik für Hämatologie und Onkologie, Eschweiler, Germany, 9Katholisches Krankenhaus, Hagen, Germany, 10Evangelisches Krankenhaus, Hamm, Germany, 11 Onkologie Leer-Emden-Papenburg, Leer, Germany, 12HämatologischOnkologische Gemeinschaftspraxis, Leipzig, Germany, 13Schwerpunktpraxis für Hämatologie und Onkologie, Magdeburg, Germany, 14Krankenhaus Barmherzige Brüder, Klinik für Onkologie und Hämatologie, Regensburg, Germany, 15Diakonie-Klinikum Stuttgart, Hämatologie und Onkologie, Stuttgart, Germany, 16Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Ludwig-Maximilians-Universität, München, Germany 1

Introduction: The CML V (TIGER)-study is a multicenter, randomized Phase III trial to evaluate efficacy and tolerability of nilotinib 300 mg twice daily (BID) monotherapy vs. nilotinib 300 mg BID + pegylated interferon alpha (PEG-IFNα2b) with the option to discontinue treatment after a nilotinib or PEG-IFNα2b maintenance therapy, respectively. A pilot phase preceded the main phase of the study to evaluate tolerability of 300 mg nilotinib BID combined with PEG-IFNα2b (30–50 µg/week). Methods: Pilot phase patients (pts) were treated with the combination of 300 mg nilotinib BID and PEG-IFNα2b (commenced after at least 6 weeks nilotinib therapy). PEG-IFNα2b was administered once weekly by s.c. injections at a dose of 30 to 50 µg/week. Treatment status, adverse events (AEs) and molecular response were analyzed for the first 12 months of therapy. Results: Between August 2012 and January 2013, 25 newly diagnosed chronic phase CML pts (7 female) were enrolled. Median age was 46 (21–74) years, 3 pts showed a high EUTOS score. All pts are alive after a median observation time of 26 (4–32) months. Nilotinib at 300 mg BID was continued in 16/25 pts, temporarily discontinued or dose-reduced in 5/25 pts and switched to another TKI due to AEs in 2/25 pts. PEG-IFNα2b was continued in 13/25 pts with a median administered dose of 30 (29–50) µg/week and median treatment duration of 45 (27–51) weeks in the first year. 3/25 pts discontinued PEG-IFNα2b temporarily. In 4/25 pts PEG-IFNα2b could not be started and in 3/25 pts PEG-IFNα2b had to be stopped permanently due to AEs. 2/25 pts dropped out of the study after withdrawal of consent. There were no pts with hematologic CTC grade ≥3 AEs. Grade ≥3 nonhematologic (mainly biochemical) AEs occurred in 6 pts under treatment with nilotinib and in 1 pt under combination therapy. A complete hematologic remission was obtained by all pts after 20–92 days (median 40 days). 17/23 pts (74%) achieved at least a major molecular response (MMR) with a proportion of deep molecular responses (≥MR4) of 57% (13/23 pts) at 12 months after randomization. Conclusions: A first line treatment with nilotinib at 300 mg BID combined with PEG-IFNα2b dosed at 30–50 µg/week and commenced after at least 6 weeks of prior nilotinib therapy is feasible for most pts and shows high response rates. Major side effects consist of skin rash, headache, fa-

Abstracts

Disclosure: No conflict of interest disclosed.

MDS klinisch II V606

Assessment of the myelodysplasia (MDS) international prognostic scoring system (IPSS) in routine care: Data from the regular care MDS registry Steinmetz T.H.1, Böttger I.1, Sauer A.2, Lathan B.3, Germing U.4, Schmitz S.1 Praxis für Hämatologie und Onkologie, Köln, Germany, 2Praxis für Hämatologie und Onkologie, Potsdam, Germany, 3Praxis für Hämatologie und Onkologie, Dortmund, Germany, 4Universitätsklinik Düsseldorf, Hämatologie und Onkologie, Düsseldorf, Germany 1

The international prognostic scoring system (IPSS) should be applied to all patients (pts) with myelodysplastic syndrome (MDS) to assess prognosis and define the optimal therapeutic strategy. The aim of the current evaluation of the basic and regular care MDS-registry was to describe the frequency, impediments and difficulties applying the IPSS in routine care. Methods: 1689 pts from 80 institutions were documented in the registry from July 2009 to March 2015 (69 months), if a bone marrow (BM) biopsy was performed and pts have given written informed consent. IPSS was documented by the centers (doc-IPSS) and in addition calculated by the documentation system (cal-IPSS), if all parameters were available. Discrepancies between cal- and doc-IPSS were queried. Results: IPSS was documented in 684 (40.5%) and calculated in 965 (57.1%) of 1689 pts. One of both was available in 1169 (69.2%) pts and both together in 480 (28.4%) pts. In 105/480 (21.9%) there was a difference between cal- and doc-IPSS. In 57/480 (11.9%) pts doc-IPSS was better than cal-IPSS, in 48/480 (10%) it was worse, respectively. The table shows the frequencies of measurement and abnormal values of the IPSS-parameters. The results of the queries will be shown in the final analysis. Tab. 1. IPSS parameters of 1689 MDS-pts (BM-biopsy in all

Parameter (abnormal)

N (%) measurement

N (%) abnormal

Hemoglobin (<10g/dl)

1653 (97.9%)

783 (47.4%)

Granulocytes (< 1.800/µL)

1472 (87.2%)

673 (45.7%)

Thrombocytes (< 100.000/ µL)

1608 (95.2%)

619 (38.5%)

BM-blasts (<5%)

1396 (82.7%)

876 (62.8%)

BM-blasts (5–9%)

1396 (82.7%)

263 (18.8%)

BM-blasts (10–19%)

1396 (82.7%)

194 (13.9%)

BM-blasts (20–30%)

1396 (82.7%)

53 (3.8%)

BM-blasts (>30%)

1396 (82.7%)

10 (0.7%)

Cytogenetic analysis (aberration)

1236 (73.2%)

489 (39.6%)

Conclusion: Despite BM-biopsies were performed in all pts, IPSS can be determined just in about 70% due to various reasons. There seem to be various uncertainties and impediments calculating IPSS in routine care. The main reasons for the lack of IPSS were missing cytogenetics and ANC counts. The MDS-registry is supported by a grant from Celgene and Novartis. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

183


CONTENTS AUTHOR INDEX

V607

Focus on humoral adaptive immunity in GATA2 haploinsufficiency Wehr C.1,2, Grotius K.1, Janda A.1,3, Wlodarski M.4, Mejstrikova E.5, Dräger R.1, Warnatz K.1, Rizzi M.1, Salzer U.1,6 Center for Chronic Immunodeficiency/ University Medical Center, Freiburg, Germany, 2Department of Hematology and Oncology, University Medical Center, Freiburg, Germany, 3Clinical Division of Pediatric Infectious Diseases and Rheumatology, Center of Pediatric and Adolescent Medicine, University of Freiburg, Freiburg, Germany, 4Division of Pediatric Hematology-Oncology, Center of Pediatric and Adolescent Medicine, University of Freiburg, Freiburg, Germany, 5Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic, 6Department of Rheumatology and Clinical Immunology, University Medical Center, Freiburg, Germany 1

Introduction: Heterozygous germline mutations in the transcription factor GATA2 result in a complex and variable clinical phenotype: the most prevalent findings are myelodysplastic syndrome, syndromal aspects (deafness, lymphedema) and immunodeficiency with an increased susceptibility to infections. In the peripheral blood a reduction of monocytes, T, B, NK and dendritic cells can be found. Paradoxically, B-cell numbers can be severely reduced while antibody levels and vaccination responses are usually not compromised, but comprehensive studies of the B-cell compartment are still missing. Patients and methods: Seventeen patients with proven mutations in GATA2 underwent detailed immunological analyses of the blood and bone marrow lymphocyte compartment. Immunoglobulin levels and vaccination titers were documented. GATA2 expression was quantified by RT-PCR in sorted B cell subpopulations derived from bone marrow and peripheral blood of healthy donors. Results: Lymphopenia resulting from a reduction in NK, B and CD4+ T cells was present in the majority of patients. B cell phenotyping revealed a consistent, profound reduction of transitional B cells and an expansion of CD21low B cells. In the bone marrow absence of CD10+ B cell precursors and a reduction of CD34+ stem cells was noted. Immunoglobulin levels were low (2/7 patients), normal (3/7) or elevated (2/7) and vaccination responses to peptides were unremarkable in 6/6 patients. Vaccination response to polysaccharides was absent in 1/7 patients. IgG subclass distribution was skewed in all patients available for this analysis. Quantitative RT-PCR analysis in sorted B cell subpopulations from healthy donors revealed GATA2 expression only in pro-B cells. Conclusion: Despite marked B cell cytopenia, immunoglobulin levels are largely maintained in GATA2 deficient patients, although minor deficiencies may occur occasionally. Lack of GATA2 expression beyond the pro B-cell stage implies that GATA2 does not play a B-cell intrinsic role during differentiation at later stages. Disclosure: No conflict of interest disclosed. V608

Systematic analysis of the period from first appearance of documented cytopenias to diagnosis of myelodysplastic syndromes – a 2-year update Schumann C.1, Hofmann W.-K.1, Nolte F.1,2 University Hospital, Mannheim, Department of Hematology and Oncology, Mannheim, Germany, 2St. Hedwig Hospital, Department of Internal Medicine, Berlin, Germany 1

Introduction: Myelodysplastic syndromes (MDS) are oligoclonal myeloid hematopoietic disorders characterized by ineffective hematopoiesis leading to peripheral cytopenias. Due to the development of an oligoclonal hematopoiesis with increasing age and the chronic nature particularly of low risk MDS it can be assumed, that MDS is already present but undiagnosed before patients become symptomatic. Therefore, we present extended and updated data showing that altered hematopoietic parameters were altered years before the diagnosis of MDS is established.

184

Oncol Res Treat 2015;38(suppl 5):1–270

Methods: We retrospectively reviewed the charts of 40 patients with MDS (male n = 22, female n = 18) at our department. Blood counts, mean corpuscular volume (MCV) and levels of lactate dehydrogenase (LDH) were evaluated from the earliest time point prior to diagnosis of MDS. Results: The median time from first detection of abnormal blood counts to diagnosis of MDS was 4.3 years (range: 1.0–34.21). In 64% of the patients hemoglobin levels were decreased as were hematocrit in 45%, erythrocytes in 81%, platelets in 36%, and white blood cells in 30% of the patients, whereas MCV was increased in 37% and LDH in 19% of the patients. MDS subtypes according to WHO classification (2008) at the time point of first diagnosis were as follows: RCMD 41%, RAEB-1 18%, RAEB-2 10%, RARS 10%, MDS-U 3%, RA 3%, 5q- 5%, and AML 3%. Both CMML-1 and CMML-2 were diagnosed in 5% of the patients, respectively. Interestingly, time of preceding hematopoietic changes did not correlate to any MDS subtype or risk category. Conclusion: Our data from this single center analysis indicate that signs of insufficient hematopoiesis might be present years before the diagnosis of MDS is established. In our cohort median time from first detection of abnormal laboratory tests to first diagnosis was 4.3 years. Interestingly, the duration of preceding hematopoietic changes did not correlate to any MDS subtype or risk category. Since several treatment approaches in MDS have shown a higher efficacy when initiated early in the course of the disease, we suggest that a consequent diagnostic work-up including cytomorphology, as well as cytogenetic and molecular genetic analyses should not be delayed in case of unexplained cytopenias. Disclosure: No conflict of interest disclosed. V609

Granulocyte and granulocyte-macrophage colony stimulating factors for patients with myelodysplastic syndromes: systematic review with meta-analysis Hutzschenreuter F.1, Kreuzer K.-A.2, Monsef I.1, Engert A.2, Skoetz N.1 Uniklinik Köln, Evidenz-basierte Onkologie, Klinik I für Innere Medizin, Köln, Germany, 2Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany 1

Introduction: Myelodysplastic syndromes (MDS) are a heterogeneous group of haematological diseases which are characterised by a dysplasia of haematological stem cells. Standard treatment is supportive care of the symptoms including red blood cell transfusions, administration of erythropoiesis stimulating agents in the case of anaemia or treatment with granulocyte and granulocyte-macrophage colony stimulating factors (G-CSF and GM-CSF) in cases of neutropenia. The objective of this review is to assess and summarise the evidence for the treatment of MDS patients with G-CSF and GM-CSF in addition to standard therapy. Methods: We searched Cochrane Central Register of Controlled Trials and MEDLINE (1950 to November 2014) for randomised controlled trials (RCTs) and included trials evaluating G-CSF or GM-CSF in addition to standard therapy in comparison to standard therapy only in MDS patients. Two review authors independently screened search results, extracted data and assessed the quality of trials. We pooled trials using the random-effects model. Results: Seven RCTs involving 569 patients were identified, however, one study presented only cumulative results for both arms. In the other six trials, data were not reported in a comparable way and patient-related outcomes like survival, time to progression to AML or incidence of infections were reported in two trials only. Five RCTs (N = 337) assessed the efficacy of G-CSF in combination with standard therapy. Meta-analyses could not be performed due to inconsistent reporting of data. One trial (N = 64) reported significantly shorter duration of neutropenia in the G-CSF + chemotherapy arm compared to the chemotherapy only arm (P = 0.015). Another trial (N = 103) found a significantly higher increase in neutrophils in the G-CSF group (P = 0.0001). Other outcomes either did not significantly differ or were not reported at all.Two RCTs (N = 232) evaluated GM-CSF in addition to standard therapy. Meta-analysis could be performed for complete response and showed no evidence for a differ-

Abstracts


CONTENTS AUTHOR INDEX

ence between both arms (RR 0.71, 95% CI 0.38 to 1.35 P = 0.76). Other outcomes did either not significantly differ or were not reported at all. Conclusions: Although seven trials have been published, this systematic review shows that there is a substantial lack of data, which would rectify the use of G-CSF and GM-CSF for the prevention of infections, prolonging of survival and improvement of quality of life. Disclosure: No conflict of interest disclosed.

V611

Gfi1 as a novel prognostic marker and for myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) and offers a novel personalized treatment Botezatu L.1, Hönes J.M.1, Michel L.1, Thiede C.2, van der Reijden B.3, Heuser M.4, Makishima H.5, Thomas R.5, Jaroslaw M.5, Ehninger G.6, Moroy T.5, Dührsen U.1, Khandanpour C.1 University Hospital Essen, Department of Hematology, Essen, Germany, University Hospital TU Dresden, Department of Molecular Hematology, Dresden, Germany, 3Radboud University Nijmegen Medical Centre, Department of Laboratory Medicine, Nijmegen, Netherlands, 4Hannover Medical School, Hannover, Germany, 5Cleveland Clinic, Cleveland, United States, 6University Hospital TU Dresden, Department of Internal Medicine I, Dresden, Germany 1

V610

Role of TP53 mutational status in MDS and AML with complex chromosomal abnormalities Schaab R.1, Ganster C.1, Rietkötter E.1, Dierks S.1, Shirneshan K.1, Haase D.1 Universitätsmedizin Göttingen, Klinik für Hämatologie und Onkologie, Göttingen, Germany 1

Introduction: In myelodysplastic syndromes (MDS) TP53 mutations of different kind occur in patients with a complex karyotype. Up to now it is unclear if the mutational status (hetero- vs. homozygous, chromosomal vs. molecular) of TP53 influences the extent of (cyto)genetic instability and the overall survival. Methods: We identified 26 patients with MDS (17), AML after MDS (2) and AML (7) with complex karyotypes (≥3 cytogenetic aberrations). Chromosomal banding (CBA), TP53-flourescence in situ hybridization (FISH), multicolor FISH and SNP-array analysis (SNP-A) were used to characterize the patients comprehensively. We compared the size of total genomic aberrations (TGA, a method to objectify the extent of genetic damage) using SNP-A and the number of cytogenetic aberrations (CA) determined by CBA in patients with and without TP53 mutations. Results: 18/26 (69%) patients showed a molecular mutation in TP53. In 12/26 (46%) patients a deletion of the short arm of chromosome 17, which involved the locus of TP53, was found by FISH/CBA. The aberration (chromosomal or molecular) of only one allele (heterozygous state) of TP53 was found in 14/26 patients (54%), two aberrations (homozygous) were found in 7/26 patients (27%). TGA size of patients with molecular TP53 mutations was significantly larger than the TGA size of those without mutations (362 Mb (97–981 Mb) vs. 185 Mb (12– 339Mb), p = 0.02). CA in patients with molecular TP53 mutations was also significantly larger compared to patients without such mutations. (7,5 (4–23) vs. 4,5 (3–11), p = 0,04). We found a positive correlation of the mutational status of TP53 with TGA size and CA: Patients with homozygous TP53 mutation showed a TGA of 464Mb (306– 981 Mb) and a CA of 12 (4–23), patients with heterozygous TP53 had a TGA of 275 Mb (12–495 Mb) and a CA of 5 (3–12). Patients with unmutated TP53 showed a TGA of 163 Mb (105–339 Mb) and a CA of 6 (3–11); p = 0,15 (TGA) and 0,10 (CA). Conclusion: So far, we could see a significant difference between the patients with and without TP53 mutations in the extent of genetic instability. 4/26 (15%) patients with complex karyotype showed no TP53 mutation at all, raising the question whether other genes of the TP53 pathway might be involved in these cases causing the same “TP53-like” phenotype. To improve our database we will continue to include patients in our project.

2

Transcription factors (TFs) are important in the onset and progression of AML. Expression data revealed that the level of the hematopoietic TF Growth factor independence 1 (GFI1) is predictive for AML development, progression and relapse. In MDS, deletion of Gfi1 and low level of Gfi1 were associated with disease progression to AML. We investigated how deletion of Gfi1 or lower expression of Gfi1 influenced disease progression and therapeutic interventions. To this end, we used mice expressing Gfi1 at different levels: mice heterozygous for Gfi1, in which one allele of Gfi1 is deleted (Gfi1het), or mice expressing Gfi1 at 20% of normal expression level (Gfi1KD) crossed with Nup98HoxD13 mice recapitulating human MDS. A shortened latency and an increased incidence of AML was found for both Gfi1KD and Gfi1het mice (p = 0.01 and p = 0.005, respectively). We confirmed our findings in an AML mouse model expressing the human oncofusion protein MLL-AF9. Gfi1 represses its target genes by recruiting histone modifying enzymes such as histone deacetylases (HDACs). We hypothesized that low levels of Gfi1 are not sufficient to initiate these epigenetic changes. Indeed, we observed increased H3K9 acetylation in Gfi1KD leukemic and preleukemic murine cells. Similarly, in GFI1 low expressing MDS patients a derepression of GFI1 target genes and failure of a HDAC signature was observed. Some MDS patients are treated with HDAC inhibitors (HDACi). We predict that treatment of AML patients with low GFI1 levels with HDACi would be ineffective and treatment with histone acetyltransferase inhibitors (HATi) more beneficial. To test this, murine leukemic cells as well as human AML cell lines expressing Gfi1 at different levels were treated with HDACi or HATi. Whereas high Gfi1 expressing murine or human leukemic cells responded to HDACi treatment, low expressing leukemic cells did not, but responded to HATi. Thus, low level of Gfi1 predisposes to AML progression by inability to repress oncogenes such as HoxA9 and this can be reversed by treatment with HATi. If low Gfi1 expression predisposes to AML progression then very high Gfi1 level should inhibit AML development. Indeed, enforced overexpression of Gfi1 in leukemic cells in vitro or in vivo hampered AML progression inducing terminal differentiation of leukemic cells. In summary, our results indicate that HATi or Gfi1 overexpression could be a novel therapeutic approach in MDS and AML patients with low GFI1 expression levels. Disclosure: No conflict of interest disclosed.

Disclosure: No conflict of interest disclosed.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

185


CONTENTS AUTHOR INDEX

Freier Vortrag

V613

Kopf-Hals-Tumore II V612

Systemic treatment pattern in a large cohort of recurrent/ metastatic head and neck squamous cell cancer (r/m HNSCC) patients (pts) treated at a tertiary cancer centre Siano M.1,2, Desax M.C.1, Früh M.1, Joerger M.1 Cantonal Hospital St. Gallen, Clinic of Oncology / Hematology, St. Gallen, Switzerland, 2Fondazione IRCCS Istituto Nazionale dei Tumori, Head and Neck Medical Oncology Unit, Milan, Italy 1

Background: HNSCC pts are often treated with systemic agents. Only one combination regimen including an anti-EGFR antibody (EAB) showed a survival benefit in a phase III trial in selected patients. Real life data are scarce. Material and methods: We retrospectively analyzed our head&neck database selecting patients with r/m HNSCC, referred to our clinic for evaluation of systemic treatment during the last 10 years. Primary study enpoint is survival. Patient, disease and treatment characteristics were assessed in all patients. Results: We included 112pts in our analysis. Mean age was 61years (range 38–85). 84% of pts were male and 16% female. 80% had an ECOG PS of 0 or 1. Oropharynx, oral cavity, larynx and hypopharynx were the primary cancer sites in 29, 26, 20 and 16% respectively, with 9% other sites (nasal cavity, rhinopharynx). 12pts(10.7%) did not receive systemic anticancer treatment due to other reasons. 100pts received first-line therapy; 83pts received a doublet combination chemotherapy containing a platinum compound(DT) and 17pts a triplet with an EAB in addition (TT). Preferred chemotherapy combination was carboplatin/paclitaxel in 62pts, combined with cetuximab in 15pts. Only 7pts received mono-therapy with carboplatin, cetuximab or paclitaxel and 3pts a non-platinum combination chemotherapy. Cetuximab with a non-platinum compound was administered only in 3pts. In total, 82pts were assessable for response. Response rate (PR, CR) was 39% with 38% having stable disease (SD). TT showed a significantly higher radiological response rate compared to DT (59 vs. 30%), p < 0.001, Fisher’s exact). CR was present in 4%(3pts) and was durable >6months. 53pts received further 2nd line treatment: 20pts a combination therapy (14pts with EAB) and 33pts a mono-therapy (25pts with EAB). In 2nd line, RR was 23% with 32% having SD. 13 out of 110pts received third line treatment with only two responses. In total 74pts received an EAB during their disease. Median survival from start of palliative systemic treatment was 8.0months (95% CI, 6.5–12.0months). One-year survival was similar in pts receiving 1st-line TT compared to DT (47% vs. 45%). ECOG performance status was the only significant prognostic parameter within multivariate Cox regression analysis (HR = 2.55, p < 0.001). Conclusion: In our single center experience similar survival compared to phase III data was achieved. 2nd line systemic treatment is active in these pts and should be considered. Disclosure: No conflict of interest disclosed.

Exploratory analysis of outcomes for patients with locally advanced or metastatic radioactive iodine-refractory differentiated thyroid cancer (RAI-rDTC) receiving open-label sorafenib post-progression on the phase III DECISION trial Paschke R.1, Schlumberger M.2, Nutting C.3, Jarzab B.4, Elisei R.5, Siena S.6, Bastholt L.7, de la Fouchardiere C.8, Pacini F.9, Shong Y.K.10, Sherman S.I.11, Smit J.W.12, Chung J.13, Kappeler C.14, Molnar I.13, Brose M.S.15 Leipzig University, Department for Endocrinology and Nephrology, Leipzig, Germany, 2Gustave Roussy, Villejuif, France, 3Royal Marsden Hospital, London, United Kingdom, 4Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Gliwice, Poland, 5University of Pisa, Department of Clinical and Experimental Medicine, Pisa, Italy, 6Ospedale Niguarda Ca’ Granda, Milan, Italy, 7Odense University Hospital, Odense, Denmark, 8Hospices Civils-Centre Anticancéreux, Consortium Cancer Thyroïdien, Lyon, France, 9 University of Siena, Unit of Endocrinology, Siena, Italy, 10University of Ulsan College of Medicine, Asan Medicine Center, Seoul, Korea, Republic of, 11The University of Texas, MD Anderson Cancer Center, Houston, United States, 12 Radboud University Medical Center, Department of Internal Medicine, Nijmegen, Netherlands, 13Bayer HealthCare Pharmaceuticals, Whippany, United States, 14Bayer Pharma AG, Berlin, Germany, 15Abramson Cancer Center of the University of Pennsylvania, Department of Otorhinolaryngology, Head and Neck Surgery, Philadelphia, United States 1

Background: RAI-rDTC patients treated with sorafenib (SOR) in the phase III placebo (PLC)-controlled DECISION trial had significantly improved progression-free survival (PFS). Patients who progressed in the double-blind (DB) period were unblinded and at the investigator’s discretion allowed to receive open-label (OL) SOR and then followed for subsequent disease progression (PFS2). Methods: PFS2, an exploratory endpoint, was defined as time from new baseline until centrally assessed progression or death during or after OLSOR treatment. Results: A total of 207 patients were randomized to sorafenib (DB-SOR) and 210 to placebo (DB-PLC). 150 PLC patients crossed over to OL-SOR at progression; 137 evaluable for efficacy (PLC-SOR). 55 DB-SOR patients continued OL-SOR at progression; 46 evaluable for efficacy (SOR-SOR). The PLC-SOR and SOR-SOR patients had poorer risk features at enrollment compared to patients not assessed for PFS2. Partial responses in the DB-SOR and PLC-SOR patients were 12.2% and 9.5%, respectively. Among SOR-SOR patients with progression events in both periods, PFS2 was more than two-fold longer than PFS1 in 22%. The median treatment duration for SOR patients receiving DB and OL treatment was 56.9 weeks. Adverse events were similar for PLC-SOR and DB-SOR patients. For SOR-SOR patients, diarrhea and hand-foot skin reactions were reduced compared to the DB-SOR treatment period. Conclusions: PFS2 for the subset of PLC patients who crossed over to OL-SOR was longer (9.6 months) than PFS1 (5.3 months). Continuation of SOR after progression needs to be explored further. Disclosure: Ralf Paschke: Advisory Role: Bayer HealthCare, Eisai; Financing of Scientific Research: Bayer HealthCare, Eisai. Marcia Brose: Advisory Role: Bayer HealthCare Pharmaceuticals, Exelixis, Onyx Pharmaceuticals; Financing of Scientific Research: Bayer HealthCare Pharmaceuticals; Expert Testimony: Bayer HealthCare Pharmaceuticals, Exelixis, Eisai, Novartis, Roche/Genentech. V614

Survival of recurrent/metastatic head and neck squamous cell cancer (r/mHNSCC) patients (pts) treated in 1st or 2nd line with an anti-EGFR-antibody (EAB) Siano M.1,2, Desax M.C.1, Früh M.1, Joerger M.1 Cantonal Hospital St. Gallen, Clinic of Oncology / Hematology, St. Gallen, Switzerland, 2Fondazione IRCCS Istituto Nazionale dei Tumori, Head and Neck Medical Oncology Unit, Milan, Italy 1

Introduction: Only one RCT was able to show a modest survival benefit in r/mHNSCC adding an EAB to platinum-combination therapy in selected pts (EXTREME 2008), whereas another didn’t (SPECTRUM 2014).

186

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Clinically relevant treatment responses are seen with 2nd line treatment, including EABs. The optimal sequence of chemotherapy and EAB remains unlcear in patients with r/mHNSCC. Material and methods: We retrospectively compared patients with r/ mHNSCC who received 1st-line platinum-combination chemotherapy in combination with an EAB (cohort E1) versus patients receiving a sequence of 1st-line platinum-combination chemotherapy followed by 2nd-line EAB (cohort E2) from a prospective HNSCC clinical database. All patients received treatment in a single referral cancer center between 2004 and 2014. The primary study endpoint is overall survival (OS), secondary study endpoints include radiological response (RR) and progression-free survival. Survival endpoints were calculated using the method of Kaplan-Meier and multivariate cox regression models. Results: We included 18 pts in cohort E1 and 34 pts in cohort E2. Combination chemotherapy in cohort 1 included carboplatin/paclitaxel in 17 pts and cisplatin/5FU in one patient. In cohort E2, 22 pts (65%) received single-agent EAB in 2nd-line (panitumumab in 59% and cetuximab in 41%), and 12pts (35%) received a combination of an EAB and chemotherapy. RR to 1st-line therapy was 56% and 32% in cohorts E1 and E2, respectively (P = 0.33). Only 10 pts (56%) in cohort E1 received 2nd line treatment, while all 34 pts in cohort E2 received 2nd-line treatment. RR in 2nd-line was 10% and 24% in cohorts E1 and E2, respectively (P = 0.002). RR over both treatment lines was 66% and 56% in cohorts E1 and E2, respectively. Median OS was similar in cohort E1 compared to cohort E2 (14.5 vs. 12 months, logrank P = 0.30). ECOG performance status was the only prognostic factor in the multivariate cox regression model (HR = 1.74, P = 0.02). Conclusion: This retrospective analysis suggests that systemic treatment with an EAB in patients with r/mHNSCC may be transferred to 2nd line without hampering clinical outcome. Disclosure: No conflict of interest disclosed. V615

One-step immediate reconstruction of the midface with patient specific PEEK implants: Challenges and chances Thieringer F.M.1, Maurer J.1, Beinemann J.1, Zeilhofer H.-F.1, Jaquiéry C.1, Kunz C.1 Universitätsspital Basel, Mund-, Kiefer- und Gesichtschirurgie, Basel, Switzerland

1

Introduction: The functional and esthetic immediate reconstruction of the midface after tumor resection still represents a major challenge for the surgeon. For an instant orofacial rehabilitation the reconstruction by autologous tissue transfer often is not the first choice of treatment due to the complex anatomical situation, prolonged onerous interventions, donor site morbidity and uncertain resection margins. Multiple step procedures can be associated with considerable functional and esthetic loss. Methods: Using the three dimensional computer-based reconstruction of the patient’s CT image data, mirroring techniques and other computer-aided design procedures an individualized PEEK (polyetheretherketone) implant of the planned midfacial resection area is produced preoperatively. In this patient specific implant (PSI) retention elements for a modified maxillary obturator prosthesis are integrated. During the same surgical procedure after tumor resection the implant can be precisely fixated to the osseous margins of the midface by titanium plates and screws. Subsequently, the modified obturator prosthesis will be incorporated. Results: In the last five years so treated patients had an unremarkable wound healing with low complication rates at our center. Operation times can be significantly shortened. This novel surgical technique for immediate maxillary reconstruction has proven to be an alternative method of choice to provide a quick recovery of orofacial functions like chewing, swallowing, speech as well as facial and dental aesthetics. Conclusions: The immediate reconstruction of the maxilla for particular indications by PSI as a temporary or final solution is possible. Patient individualized prefabricated PEEK implants with appropriate obturator prosthesis for maxillary replacement are promising variants of a functional and esthetic rehabilitation. Disclosure: No conflict of interest disclosed.

Abstracts

Wissenschaftliches Symposium

Young Investigator‘s Award Preisverleihung V616

The Serine-Threonine Kinase 36 regulates growth, metastasis, differentiation and cancer stem cell maintenance of solid tumors by modifying Hedgehog, Wnt and Notch signaling Benkler T.1, Höfler S.1, Kuhn A.1, Pfeifer D.1, Follo M.1, Timme-Bronsert S.2, Lassmann S.2, Brummer T.3, Arnold S.4, Duyster J.1, Dierks C.1 Uniklinik Freiburg, Innere Medizin, Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 2Uniklinik Freiburg, Institut für Pathologie, Freiburg, Germany, 3Albert-Ludwigs-Universität Freiburg, Institut für Molekulare Medizin und Zellforschung, Freiburg, Germany, 4Uniklinik Freiburg, Zentrale Klinische Forschung, Freiburg, Germany 1

The Serine-Threonine Kinase 36 is part of the Hedgehog signaling complex downstream of the SMO- and PTCH receptors and transmits the activation and nuclear localization of the GLI transcription factors. In a siRNA kinase screen of 30 tumor cell lines, we found that STK36 is important for tumor cell growth especially in pancreatic cancer (n = 5), ovarian cancer (n = 3) and melanoma cell lines (n = 4), while the growth of glioblastoma or medulloblastoma cell lines was not affected. STK36kd responsive samples were mainly resistant towards kd of the upstream SMO receptor, implicating a downstream activation mechanism. IHC staining of STK36 in primary tumor samples from ovarian cancer patients (n = 40) and pancreatic cancer patients (n = 25) showed a strong overexpression and positive correlation with GLI1 expression and nuclear localization, implicating STK36 overexpression as an alternative HH-activation mechanism. Inducible knockdown of STK36 with two different shRNA sequences revealed a strong reduction of proliferation and especially cancer cell migration and tumor invasion. In agreement, overexpression of STK36 in non-malignant ovarian epithelial cells enhanced proliferation and cell migration. In vivo knockdown of STK36 confirmed our results, showing reduced tumor growth and a nearly complete abrogation of tumor metastasis. Most interestingly, we found that the knockdown of STK36 in pancreatic cancer cells (MiaPaCa2) and melanoma cells (CHL1) caused differentiation of primary tumors and reduced the cancer stem cell frequency. This effect could be reconfirmed in vitro, where the knockdown of STK36 abrogated the ability of cancer cells (MiaPaCa2, CHL1) to form spheroids in 3D-culture assays, while cell line (ES-2) which does not form spheroids was not affected. Gene expression analysis revealed, that knockdown of STK36 not only reduced the activity of the HH signaling pathway, but even more reduced Notch and Wnt signaling, all major pathways important for stem cell maintenance and differentiation. On biochemical level, the knockdown of STK36 reduced the nuclear translocation of the transcriptional factor GLI3. In conclusion, our results demonstrate overexpression of STK36 as a potential alternative activation mechanism of HH-signaling in cancer cells downstream of the SMO receptor, affecting tumor growth, tumor metastasis, tumor differentiation and cancer stem cell maintenance by modifying essential stemness pathways, thus implicating STK36 as a novel drug target. Disclosure: No conflict of interest disclosed. V617

NIPA as a novel regulator of aging and stress response of the primitive HSC pool Kreutmair S.1, Istvanffy R.2, Klingeberg C.1, Dierks C.1, Oostendorp R.2, Duyster J.1,3, Illert A.L.1,3 Universitätsklinikum, Freiburg, Germany, 2Klinikum rechts der Isar der Technischen Universität, München, Germany, 3German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany 1

Accumulation of DNA damage in hematopoietic stem cells (HSCs) is associated with aging, bone marrow failure and development of hematological malignancies. Although HSCs numerically expand with age, their

Oncol Res Treat 2015;38(suppl 5):1–270

187


CONTENTS AUTHOR INDEX

functional activity declines over time and the protection mechanism from DNA damage accumulation remains to be elucidated. NIPA (Nuclear Interaction Partner of ALK) is highly expressed in hematopoietic stem and progenitor cells, especially in the most primitive long-term repopulating HSCs (CD34-Flt3-Lin-Sca1+cKit+). Loss of NIPA leads to a significant exhaustion of primitive hematopoietic cells, where Lin-Sca1+cKit+ (LSK) cells were reduced to 40% of wildtype (wt) littermates (p < 0,001). All LSK-subgroups, LT-HSCs (p < 0.001), STHSCs (CD34+Flt3-LSK; p < 0.01) and MPPs (CD34+Flt3+LSK; p < 0.05) of NIPA deficient animals are affected and failed to age-related increase, whereas the lineage differentiation of NIPA-/- progenitor cells showed no gross differences. Myeloid depression by 5-FU treatment led to severely reduced HSC self renewal in Nipa-/- mice independent of age (p < 0.001). Moreover, weekly 5-FU activation showed reduced survival of Nipa-/- vs. wt animals (11 vs. 14.5 days). To further examine the role of NIPA in HSC maintenance and exhaustion, we performed in vivo repopulation experiments, where NIPA deletion causes bone marrow failure in case of competition, as Nipa-/- cells contributed to less than 10% of transplanted BM cells 6 month after transplantation (TX). According to this, colony formation assays and limiting dilution transplantation showed a dramatic reduction of competitive repopulation units (p < 0,0001) in NIPA-/- animals. Serial LSK transplantation assays revealed loss of NIPA-deficient LSKs shortly after TX, whereas long-term repopulation capacity seemed to be maintained, suggesting a role of NIPA in critical stress response. To further investigate the stress response in NIPA-deficient HSCs, we irradiated LSKs with 3 Gy and stained for DNA-Damage foci by pH2ax. Remarkably, loss of NIPA led to significant higher numbers of pH2ax foci in irradiated HSCs (46% > 6 foci vs 17% > 6 foci in wt cells) and highly increased the rates of apoptotic cells especially in the primitive CD34-LSK population. Taken together our results highlight the importance of the DNA damage response at HSC level for lifelong hematopoiesis and establish NIPA as a novel regulator of aging and stress response of the primitive HSC pool.

Methods: A multicenter randomized controlled trial was conducted at 10 German hematology departments. We compared chlorhexidine containing highly adhesive dressings with non-chlorhexidine highly adhesive dressings (control) in high-risk neutropenic patients. The primary endpoint was the incidence of definite CRBSI within the first 14 days (dCRBSI14) of central venous catheter (CVC) placement. Secondary endpoints included overall incidence of definite CRBSI (dCRBSI), 14 days and overall incidence of definite or probable CRBSI (dpCRBSI14 and dpCRBSI) and incidence of unscheduled dressing changes and adverse events. CRBSI were defined according to the Guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO) from 2008. Results: From Feb 2012 to Sept 2014, 630 patients were included in the study. 17 patients were excluded from evaluation. Incidence of dCRBSI14 was 2.6% (8/307) in the chlorhexidine and 3.9% (12/306) in the control group (p = 0.375), and for dCRBSI 4.2% (13/307) vs. 7.9% (24/306), respectively (p = 0.064). Both dpCBRSI14 and dpCRBSI were significantly less frequent in the study group with dpCRBSI14 in 6.5% (20/307) of the chlorhexidine group as compared to 11% (34/306) in the control group (p = 0.047), and dpCRBSI in 10.4% (32/307) vs. 17% (52/306), respectively (p = 0.019). Dressing intolerance leading to discontinuation was similar between both groups (7.2% and 7.8%). Conclusion: The application of chlorhexidine containing iv-catheter securement dressings reduces the incidence of definite or probable CRBSI in neutropenic patients. Clinical trial registered with Clinicaltrials.gov (NCT01544686). Disclosure: Lena Biehl: Financing of Scientific Research: LMB has served at the speakers’ bureau of Astellas and MSD.; Expert Testimony: LMB is supported by the German centre for Infection Research (DZIF). Maria Vehreschild: Advisory Role: MJGTV is a consultant to Berlin Chemie.; Financing of Scientific Research: MJGTV has served at the speakers’ bureau of Pfizer, Merck, Gilead Sciences, and Astellas Pharma.; Expert Testimony: MJGTV has received research funding from 3M, Astellas Pharma and Gilead Sciences.

Disclosure: No conflict of interest disclosed. V619 V618

Chlorhexidine containing iv-catheter securement dressings for the prevention of central venous catheter-related bloodstream infections in neutropenic patients: A randomized trial (COAT study) Biehl L.M. , Huth A. , Panse J. , Hentrich M. , Engelhardt M. , Schäfer-Eckart K.6, Kofla G.7, Kiehl M.8, Wendtner C.9, Karthaus M.10, Hellmich M.11, Christ H.11, Cornely O.A.1,12,13, Vehreschild M.J.G.T.1,2 1,2

1

3

4

5

University Hospital of Cologne, Department I of Internal Medicine, Cologne, Germany, 2German Centre for Infection Research (DZIF), Site Bonn/Cologne, Cologne, Germany, 3RWTH Aachen University Hospital, Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Aachen, Germany, 4Red Cross Hospital Munich, Department of Medicine III, Munich, Germany, 5Medical Center – University of Freiburg, Department of Medicine I, Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 6Klinikum Nuernberg, Medical Clinic 5, Hematology and Oncology, Nuernberg, Germany, 7Charité – University Medicine Berlin, Department of Medicine, Division of Oncology/Hematology, Berlin, Germany, 8Clinical Center Frankfurt/Oder, Medical Clinic I, Hematology and Medical Oncology, Frankfurt/ Oder, Germany, 9Klinikum Schwabing, Department of Haematology, Oncology, Immunology, Palliative Care, Infectious Diseases and Tropical Medicine, Munich, Germany, 10Klinikum Neuperlach, Department of Hematology and Oncology, Munich, Germany, 11University of Cologne, Institute of Medical Statistics, Informatics and Epidemiology, Cologne, Germany, 12University of Cologne, Center for Integrated Oncology CIO Köln/Bonn, Cologne, Germany, 13University of Cologne, Cologne Excellence Cluster on Cellular Stress Responses in AgingAssociated Diseases (CECAD), Cologne, Germany 1

Introduction: Central venous catheter-related bloodstream infections (CRBSI) are a frequent cause of morbidity and mortality in patients with chemotherapy-induced neutropenia. Chlorhexidine containing iv-catheter securement dressings may prevent CRBSI.

188

Oncol Res Treat 2015;38(suppl 5):1–270

Dynamics of BCR-ABL independent gene mutations in patients with chronic myeloid leukemia Obstfelder E.1, Rinke J.1, Schmidt M.1, Schäfer V.1, Waldau A.1, Winkelmann N.1, Hochhaus A.1, Ernst T.1 Klinik für Innere Medizin II, Universitätsklinikum Jena, Hämatologie und Internistische Onkologie, Jena, Germany 1

Introduction: Living without treatment has become a realistic target for patients with chronic myeloid leukemia (CML) who achieved durable deep molecular remission under treatment with tyrosine kinase inhibitors (TKI). Recently, we have identified novel BCR-ABL-independent gene mutations in newly diagnosed CML patients that may function as important cofactors in the evolution of CML and persistence of the disease (Schmidt M, Rinke J, et al.: Leukemia 2014;28(12):2292–2299). Here, we sought to investigate the dynamics of such mutations during a 2-year treatment with the second generation TKI nilotinib. Methods: We randomly selected 15 CML patients in chronic phase (male, n = 10; median age 58 years, range 42–73 years). For each patient, samples were analyzed at diagnosis, in subsequent complete cytogenetic remission (CCyR) after 3 months and in deep molecular remission (≤ MMR) after 24 months of therapy with nilotinib. Targeted deep next-generation sequencing (NGS) was used to analyse a panel of 30 commonly mutated genes in myeloid disorders. Constitutional DNA obtained from buccal swabs at diagnosis was investigated to check whether mutations are of somatic origin. Results: BCR-ABL independent gene mutations were found in 5/15 (33%) CML patients at diagnosis affecting the genes ASXL1, DNMT3A, NOTCH1, RUNX1 and TET2. No mutation was recognized in corresponding constitutional DNA specimens indicating that all mutations were somatically acquired. Analysis of individual hematopoietic colonies revealed that most mutations were part of the Ph-positive clone. NGS of subsequent samples

Abstracts


CONTENTS AUTHOR INDEX

obtained after 3 months of TKI therapy identified one DNMT3A mutation in Ph-negative cells that was also present in Ph-positive cells at diagnosis, implying that this mutation preceded the BCR-ABL rearrangement. After 24 months of TKI treatment, NGS revealed novel mutations (EZH2, NOTCH1, and RUNX1) in three patients with deep molecular remissions (≤ MMR) that were neither detectable at diagnosis nor after 3 months of treatment indicating emergence of BCR-ABL-independent subclones in Ph-negative cells during long-term nilotinib treatment. Conclusions: BCR-ABL independent gene mutations can be found frequently in CML patients. Such molecular aberrations may function as important cofactors in the evolution of CML and persistence of the disease. Our findings may provide important novel genetic information regarding CML biology and for the design and performance of discontinuation trials. Disclosure: No conflict of interest disclosed. V620

A randomized phase-3 study in patients with chemotherapyinduced sensorimotor polyneuropathy evaluating an integrated program including massage, structured kinesthetic three-dimensional mobilization, physical exercises with or without whole-body vibration training Schönsteiner S.1, Kirchner E.2, Mack S.2, Bauder-Mißbach H.2, Hamel T.2, Fürniß T.2, Orth M.2, Landwehrmeyer B.2, Riester A.2, Prokein A.2, Erhardt E.2, Kunecki J.2, Eisenschink A.M.2, Pieger R.2, Döhner H.2, Schlenk R.F.2 Uniklinik Ulm, Klinik für Innere Medizin III, Ulm, Germany, 2Uniklinik, Ulm, Germany 1

Background: Peripheral sensorimotor polyneuropathy (PNP) frequent results from toxicity of chemotherapy and single agent IMiDs or proteasom inhibitors. Chemotherapy-induced PNP (ciPNP) is characterized by an enormous interference with daily activity of life because of pain, severe disturbance of fine motor skills and insomnia. The therapeutic benefit of medical treatment is uncertain. Methods: In a phase-III study patients with ciPNP grade 2/3 according to NCI CTC were randomized for an integrated program including massage, kinesthetic three-dimensional mobilization, and physical exercises with or without vibration platform training. All patients were intended to receive 15 sessions in a time period of 12 to 15 weeks. Patients were evaluated before, during and after treatment using locomotoric- and sensory tests, neurological evaluation and self-assessment. Results: A total of 131 patients with ciPNP were randomized either into the standard arm (n = 65, integrated program only) or the experimental arm (n = 66, integrated program with vibration platform training). The median age was 60 years (range 24–71years); n = 44 patients had haematological neoplasms (n = 19 multiple myelomas, n = 18 lymphomas, n = 7 leukemias) and n = 87 solide tumors (n = 27 colorectal, n = 8 lung, n = 8 oesophageal/gastric, n = 25 breast/ovarian, n = 19 others). All patients had prior chemotherapy and n = 46 had additional radiotherapy. At baseline, according to the visual analog scale (best, 0–10, worst) polyneuropathy-symptoms including pain were predominantly present in toes (median 8.1) and fingers (median 6.4). During the intervention time period n = 19 (standard) and n = 32 (experimental) patients left the program due to progression/relapse, concomitant disease or death. Overall, the patients experienced marked improvement with significantly less symptoms and pain (p < 0.001), better physical fitness and coordination (Esslinger-fitness-index, p = 0.006; chair-rising-test p < 0.001), and better results in quantitative sensory testing (QST) with thermal perception, blunt pressure, mechanical pain and vibration thresholds. Significant differences were noted in favor of the vibration platform training in daily walking distance within the first sessions (p < 0.001) and overall in improved thermal QST perception (p = 0.03). Conclusions: The treatment of ciPNP with an integrated program had a significant and clinically extremely relevant beneficial impact on symptom relieve, physical fitness and sensory function. Disclosure: No conflict of interest disclosed.

Abstracts

V621

The effects of the BRD4 blocker JQ1 on proliferation and survival of leukemic cells in Ph+ CML Peter B.1,2, Stefanzl G.2, Cerny-Reiterer S.1,2, Berger D.2, Zuber J.3, Valent P.1,2 Medical University of Vienna, Ludwig Boltzmann Cluster Oncology, Vienna, Austria, 2Medical University of Vienna, Department for Internal Medicine I, Division of Hematology and Hemostaseology, Vienna, Austria, 3Research Institute of Molecular Pathology (IMP), Vienna, Austria 1

Chronic myeloid leukemia (CML) is a hematopoietic stem cell disease in which BCR/ABL1 enhances growth and survival of leukemic cells. In most patients, clonal cells can be kept under control by BCR/ABL1 tyrosine kinase inhibitors (TKI) such as imatinib, nilotinib, dasatinib, bosutinib or ponatinib. Nevertheless, resistance against TKI may occur. Therefore, current research is seeking for novel targets in CML. One class of promising targets may be epigenetic regulators of cell growth. In acute myeloid leukemia (AML), the epigenetic reader BRD4 has recently been identified as a novel drug target. However, so far, little is known about the expression and function of BRD4 in CML cells. The aims of this study were to examine the expression of BRD4 in CML cells and to define whether BRD4 might serve as a drug target in this leukemia. As determined by qPCR and immunocytochemistry, the CML cell lines KU812 and K562 express BRD4 mRNA and the BRD4 protein. The BRD4-targeting drug JQ1 was found to inhibit 3H-thymidine uptake and thus proliferation in KU812 cells in a dose-dependent manner (IC50: 0.25–0.75 µM). Interestingly, however, no substantial growth-inhibitory effects of JQ1 were seen in K562 cells (IC50: >5 µM). In subsequent experiments, we were able to show that JQ1 inhibits growth of primary CML cells with IC50 values ranged between 0.1 to 5 µM. JQ1 was found to induce apoptosis in KU812 cells but did not induce apoptosis in K562 cells as determined by AnnexinV staining. Nevertheless, we were able to show that in both cell lines (KU812 and K562), JQ1 decreases the expression of MYC mRNA levels and MYC protein levels. Next, we analyzed whether MYC expression in CML cells is BCR/ ABL1-dependent. In these experiments, we found that imatinib, nilotinib, ponatinib and dasatinib counteract the expression of MYC mRNA as well as MYC protein expression in K562 cells and KU812 cells. Finally, we were able to show that JQ1 cooperates with imatinib, nilotinib, ponatinib and dasatinib in producing growth inhibition in KU812 cells and K562 cells. Together, our data show that BRD4 serves as a potential target in CML cells, and that the BRD4 blocker JQ1 synergizes with BCR/ABL1 TKI to induce growth-inhibition. Whether BRD4 inhibition is a pharmacologically meaningful approach in patients with TKI-resistant CML remains to be determined in forthcoming studies. Disclosure: Barbara Peter: No conflict of interest disclosed. Peter Valent: Financing of Scientific Research: Novartis; Ariad; Pfizer; Expert Testimony: Novartis.

Expertenseminar

Früherkennung Lungenkarzinom V622

Lung cancer screening Hoffmann H.1 Thoraxklinik, Universität, Heidelberg, Germany

1

The US National Lung Screening Trial (NLST) has clearly changed the landscape of lung cancer screening, since it provides strong evidence that LDCT screening can reduce lung cancer and all-cause mortality. In recognition of these results, the US Preventive Services Task Force in Lung cancer has recently announced its support of a recommendation for CT-based lung cancer screening in 55–80 year olds who have 30 pack years or smoking or up to 15 years as an ex-smoker , which means that private insurers in the US will be required to provide the full cost of CT-screening in this population over the next year. Moreover, all current US guidelines recommend lung cancer screening with LDCT. The National Comprehensive Cancer Network

Oncol Res Treat 2015;38(suppl 5):1–270

189


CONTENTS AUTHOR INDEX

(NCCN) Lung Cancer Screening Panel was the first to report, after reviewing the available evidence and has updated its recommendations since then. Further recommendations followed from the American Lung Association, the American College of Chest Physicians, the American Association for Thoracic Surgery, and the American Cancer Society. Nevertheless, uncertainty exists about potential harms and the generalizability of results. These concerns should be addressed before screening becomes standard care in Germany. Recently, European randomized lung cancer CT screening (EUCT) trial investigators published a position statement with regard to the implementation of CT screening and the performance of CT screening outside clinical trials in Europe. The EUCT consider that CT screening is not ready for implementation in Europe at this time and that we should await the outcome of the ongoing EU screening trials in order to answer the open questions: 1. Identification of high-risk individuals for lung cancer CT screening programs. 2. Develop radiological guidelines for use in developing national screening programs. 3. Develop guidelines for the clinical work-up of “indeterminate nodules” resulting from CT screening programs. 4. Guidelines for pathology reporting of nodules from lung cancer CT screening programs. 5. Recommendations for surgical and therapeutic interventions of suspicious nodules identified through lung cancer CT screening programs. 6. Integration of smoking cessation practices into future national lung cancer CT screening programs. These issues remain to be resolved in order to establish the most efficient lung cancer screening program in the future within our national health care system.

improvement of detection of fungal pathogens by defining the optimal use of biomarkers. Methods: Clinical -simultaneously taken- samples (bronchoalveolar lavage (BAL) and peripheral blood) of 101 immunocompromised patients with suspected IFI and haemato-oncological diseases were investigated within a multicentre prospective study (ClinicalTrials.gov Identifier: NCT01695512). Results of a Aspergillus PCR assay, a multifungal DNA microarray (DNA Chip) detecting 15 fungal species, a galactomannan EIA (GM) and a 1,3-beta-D-glucan assay (BDG) were analyzed. 15 Aspergillus DNA-positive samples were further analyzed for the four most common triazole resistance mutations in Aspergillus fumigatus by PCR assays and consecutive DNA sequence analysis. Results: According to EORTC/MSG criteria, patients were classified as proven (n = 2), probable (n = 26), possible (n = 56), and no IFI (n = 17). Table 1 shows the diagnostic performance of different test combinations for detection of IFI in proven/probable vs. no IFI pts. Tab. 1. Diagnostic performance of test combinations

Specimen

Test combination

Sensitivity, %

Speci­ ficity, %

PPV, %

NPV, %

DOR

Blood

PCR or Chip or GM or BDG

75

82

88

67

14

BAL

PCR (IA)

36

100

100

49

>200

BAL

GM (IA)

36

100

100

49

>200

BAL

BDG (IFI)

79

53

73

60

4.1

BAL

PCR or Chip or GM or BDG

89

41

71

70

5.8

BAL

PCR and Chip and GM and BDG

33

100

100

89

>200

BAL

PCR or GM (IA)

50

100

100

56

>200

Supportive Therapie I

Blood/ BAL

GM (blood) or PCR or Chip (BAL)

57

77

80

52

4.3

V625

Blood/ BAL

BDG (blood) or GM (BAL)

79

100

100

74

>200

Disclosure: No conflict of interest disclosed.

Freier Vortrag

Performance of combinations of Aspergillus PCR, multifungal DNA microarray, galactomannan, 1,3-beta-D-glucan and Aspergillus azole resistance PCRs in blood and BAL samples for diagnosis and characterization of invasive fungal infections in haematological patients Boch T.1, Spiess B.2, Cornely O.A.3, Vehreschild J.J.4, Rath P.M.5, Steinmann J.5, Heinz W.6, Hahn J.7, Krause S.W.8, Kiehl M.G.9, Egerer G.10, Koldehoff M.11, Liebregts T.11, Klein M.12, Nolte F.1, Will S.1, Merker N.1, Hofmann W.K.1, Buchheidt D.1, Reinwald M.1 Universitätsmedizin Mannheim; III. Medizinische Klinik; Hämatologie und Onkologie, Mannheim, Germany, 2Universitätsmedizin Mannheim; III. Medizinische Klinik; Hämatologie und Onkologie, Forschungslabor, Mannheim, Germany, 3Universitätsklinikum Köln; I. Medizinische Klinik; Clinical Trials Centre; ZKS; CIO; Köln CECAD; Universität Köln, Köln, Germany, 4Universitätsklinikum Köln, I. Medizinische Klinik; German Centre for Infection Research, Köln, Germany, 5Universitätsklinikum Essen; Institut für Medizinische Mikrobiologie, Essen, Germany, 6Universitätsklinikum Würzburg; II. Medizinische Klinik, Würzburg, Germany, 7Universitätsklinikum Regensburg; Hämatologie und Onkologie, Regensburg, Germany, 8Universitätsklinikum Erlangen; Hämatologie und Onkologie, Erlangen, Germany, 9Klinikum Frankfurt/Oder; I. Medizinische Klinik, Frankfurt/Oder, Germany, 10Universitätsklinikum Heidelberg; V. Medizinische Klinik, Heidelberg, Germany, 11Universitätsklinikum Essen; Abteilung für Knochenmarktransplantation, Essen, Germany, 12Prosper-Hospital Recklinghausen; I. Medizinische Klinik, Recklinghausen, Germany 1

Table Diagnostic performance of test combinations (PPV = positive predictive value; NPV = negative predictive value; DOR = diagnostic odds ratio) Analysis of the Aspergillus DNA-positive samples revealed a TR34/L98H cyp51A alteration in one patient with aplastic anemia. Conclusions: Evaluation of data of this study reveals that combining biomarkers is superior to their sole use in diagnosing IFI. Performance of biomarkers in blood alone is inferior to BAL, but integrating blood and BAL into the diagnostic algorithm is an advantageous approach. DNA microarray analysis broadens the spectrum of detectable fungal pathogens beyond Aspergillus spp. This study was supported by a scientific grant from Gilead Sciences, Germany. Disclosure: Tobias Boch: Financing of Scientific Research: MSD; Expert Testimony: Gilead Sciences. Mark Reinwald: Financing of Scientific Research: MSD; Expert Testimony: Gilead Sciences.

Introduction: High mortality rates of invasive fungal infections (IFI), especially invasive aspergillosis (IA), in immunocompromised haemato-oncological patients (pts) and current diagnostic limitations require

190

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V626

Identification of invasive fungal infections in immunocompromised patients by combining an Aspergillus specific PCR with a multifungal DNA microarray Boch T.1, Reinwald M.1, Postina P.1, Cornely O.A.2, Heußel C.P.3, Heinz W.4, Hoenigl M.5, Eigl S.5, Lehrnbecher T.6, Hahn J.7, Claus B.8, Lauten M.9, Egerer G.10, Müller M.C.1, Will S.1, Merker N.1, Hofmann W.K.1, Buchheidt D.1, Spiess B.1 Universitätsmedizin Mannheim; III. Medizinische Klinik; Hämatologie und Onkologie, Mannheim, Germany, 2Universitätsklinikum Köln; I. Medizinische Klinik; Clinical Trials Centre; ZKS; CIO; Köln CECAD; Universität Köln, Köln, Germany, 3Universitätsklinikum Heidelberg; Thoraxklinik, Heidelberg, Germany, 4 Universitätsklinikum Würzburg; II. Medizinische Klinik, Würzburg, Germany, 5 Universitätsklinikum Graz; Abteilung für Pulmonologie, Infektions- und Tropenmedizin, Graz, Austria, 6Universitätsklinikum Frankfurt; Pädiatrische Hämatologie und Onkologie, Frankfurt, Germany, 7Universitätsklinikum Regensburg; Hämatologie und Onkologie, Regensburg, Germany, 8Klinikum, Ludwigshafen, Germany, 9Universitätsklinikum Schleswig-Holstein; Campus Lübeck; Pädiatrie, Lübeck, Germany, 10Universitätsklinikum Heidelberg; V. Medizinische Klinik, Heidelberg, Germany 1

Introduction: The increasing incidence of invasive fungal infections (IFI) in immunocompromised patients emphasizes the need to improve the molecular tools for detection of the fungal pathogens, particularly as the diagnosis of fungal infections is rarely based on positive culture yield in this group of patients. Methods: We investigated an Aspergillus specific polymerase chain reaction (PCR) assay in combination with a multifungal DNA microarray detecting DNA of 15 different fungal pathogens, combining multiplex PCR and consecutive DNA chip hybridization. The array encompasses detection of Aspergillus, Candida, Fusarium, Mucor, Rhizopus, Scedosporium, and Trichosporon species. Biopsies, BAL (bronchoalveolar lavage), and blood samples of 133 immunocompromised patients (pts), mainly suffering from hematological diseases, were investigated: Mostly during antifungal therapy, with proven (n = 18), probable (n = 29), possible (n = 46) and no IFI (n = 40) according to 2008 EORTC/MSG or Mikulska/Blot et al. criteria for ICU pts. Molecular results were compared to results from culture, histopathology, imaging, and serology. Results: Genomic DNA and specific hybridization patterns of C. albicans, C. dubliniensis, C. glabrata, S. prolificans, R. microsporus, R. oryzae and M. racemosus were detected. Evaluation of the investigation of all samples using the microarray analysis yielded a sensitivity of 64% and a specificity of 80% for IFI other than IA. Best results were achieved when combining Aspergillus specific PCR and DNA microarray analysis for the investigation of biopsy samples. Here, the sensitivity was 79%, the specificity 71%. Conclusions: Our assays in combination rapidly detect and identify genomic DNA of fungal pathogens with best results in biopsies, suggesting clinical significance to improve identification of IFI, especially with regard to the diagnostic uncertainty of culture and/or histopathology findings during antifungal therapy. Disclosure: No conflict of interest disclosed. V627

Emergence of daptomycin non-susceptiblity in colonizing vancomycin-resistant Enterococcus faecium isolates during daptomycin therapy Lellek H.1, Franke G.2, Ruckert C.2, Wolters M.2, Wolschke C.3, Christner M.2, Büttner H.2, Alawi M.4, Kröger N.3, Rohde H.2 Universitätsklinikum Hamburg-Eppendorf, Klinik für Stammzelltransplantation, Hamburg, Germany, 2Medizinische Mikrobiologie, Virologie und Hygiene, Hamburg, Germany, 3Klinik für Stammzelltransplantation, Hamburg, Germany, 4 Bioinformatics Service Facility, Hamburg, Germany 1

Infections due to Vancomycin-resistant enterococci are of significant importance in high-risk populations, and daptomycin is a key front-line bactericidal antibiotic to treat multidrug-resistant VRE in these patients.

Abstracts

The emergence of daptomycin non-susceptibility in invasive VRE during daptomycin therapy is a major clinical issue. Here the hypothesis was tested that systemic daptomycin therapy also induces the emergence of daptomycin non-susceptible (DNS-) isolates in colonizing VRE populations. 11 vancomycin-resistant E. faecium strain pairs recovered from rectal swabs were available for analysis. All initial isolates exhibited daptomycin MICs within the wild type MIC distribution of E. faecium (MIC ≤ 4 mg/L). In follow-up isolates from five patients a 4–16-fold daptomycin MIC increase was detected. All patients carrying DNS-VRE received daptomycin (14–28 days) at 4 mg/kg body weight, while only one patient in whom no DNSE emerged was treated with daptomycin for 2 days. Comparative whole genome sequencing identified DNS-VRE-specific single nucleotide polymorphisms (SNP), including mutations in cardiolipin synthase (Cls), and additional SNPs in independent genes potentially relevant for the DNS phenotype. Mutations within cls were also identified in three additional, colonizing DNS-VRE. Of these, at least one strain was transmitted within the hospital. In no VRE pre-existing or novel mutations in the liaFSR operon were detected. This is the first report documenting the emergence of DNS-VRE in colonizing strains during daptomycin treatment, putting the patient at risk for subsequent DNS-VRE infections and priming the spread of DNS-VRE within the hospital environment. Disclosure: No conflict of interest disclosed. V628

Delayed entry of blood cultures into blood culture incubators has no negative effect of on overall culture positivity Schalk E.1, Färber J.2, Fischer T.1 Otto-von-Guericke University Magdeburg, Medical Centre, Department of Haematology and Oncology, Magdeburg, Germany, 2Otto-von-Guericke University Magdeburg, Medical Centre, Department of Medical Microbiology, Infection Control and Prevention, Magdeburg, Germany 1

Introduction: Blood cultures (BC) are an important tool for microbiological diagnostics of bloodstream infections. It is recommended that incubation of blood bottles should occur within a short period of time after collection, because of the increased risk of false-negative BC after a delay of more than 12 h. However, there are no experimental data whether delayed incubation affects the time-to-positivity (TTP) of BC. Particularly for laboratories with no 24 h service at 7 days per week, insufficient pathogen detection may negatively impact on morbidity and mortality of patients, especially in febrile neutropenia. Methods: Blood from healthy donors was drawn in 7 mL Na-Heparin vacutainers and was warmed up to 38.3°C. The prepared blood was transferred in BACTEC™ Plus Aerobic/F, Anaerobic/F and Lytic 10 Anaerobic/F and Mycosis-IC/F Culture Vials and incubated in BACTEC™ 9120 or 9240 BC automats (Becton Dickinson, Heidelberg, Germany) followed by immediate incubation with Staphylococcus aureus, S. epidermidis, Escherichia coli, Pseudomonas aeruginosa, Streptococcus mitis, Enterococcus faecium and Candida albicans (ATCC™ standard pathogens), respectively, at a concentration of 5 CFU/mL. After inoculation with blood and pathogens, the bottles were stored at room temperature and were then transferred into the incubator with a delay of 0, 2, 4, 8, 12 and 16 h. Each experiment was repeated three times for each pathogen and time point of delayed incubation. Results: Overall, n = 520 BC bottles were investigated. Of note, using Aerobic or Lytic Anaerobic bottles there were no significant changes in TTP when comparing time point 0 h with time points of delay (2, 4, 8, 12 and 16 h). Interestingly, delayed entry changed the TTP counterintuitively when comparing time points 0 h with 12 h and 16 h, respectively, in Anaerobic bottles. Thus, TTP was reduced by 7.69 h (p = 0.006) and by 6.38 h (p = 0.01), respectively. Additionally, false-negative findings were frequently observed for P. aeruginosa and C. albicans in Lytic Anaerobic bottles. Our data also revealed that the Mycosis bottle is absolutely required for early detection of C. albicans, since TTP of Mycosis bottles compared with Anaerobic bottles was shortened by 17.37 h (p = 0.0004).

Oncol Res Treat 2015;38(suppl 5):1–270

191


CONTENTS AUTHOR INDEX

Conclusions: Delayed entry of blood culture bottles into blood culture systems has no significant effect with regard to time-to-positivity in this ex vivo analysis modelling the situation of a febrile patient. Disclosure: Enrico Schalk: Expert Testimony: ES received research support from Becton Dickinson with in part free reagents. Thomas Fischer: No conflict of interest disclosed. V629

Adherence to antiemetic guidelines for highly emetogenic chemotherapy – a retrospective representative sample survey and analysis in Germany Link H.1, Nietsch J.2, Kerkmann M.2, Jordan K.3, Ortner P.4, ASORS / AIO Deutsche Krebsgesellschaft Westpfalz-Klinikum, Klinik für Innere Medizin I, Kaiserslautern, Germany, 2MMF GmbH, Dortmund, Germany, 3Universitätsklinikum Halle, Klinik für Innere Medizin IV, Halle, Germany, 4Pomme-med GmbH, München, Germany 1

Introduction: Antiemetic guidelines (GL) of ASCO and MASCC/ESMO recommend for highly emetogenic chemotherapy (HEC, emetic risk > 90%) a prophylaxis with a 5-HT3-receptor antagonist (5HT3RA), NK1-receptor antagonist (NK1RA) and dexamethasone (DEX) in the acute phase (first 24h). In the delayed phase DEX should be combined with NK1RA for 2 days (if not given i.v. on d1). In anthracycline and cyclophosphamide (AC) chemotherapy a NK1RA with DEX (ASCO) or without DEX (MASCC) is recommended in the delayed phase. Methods: Patients (pts) treated with 1–4 cycles of cisplatin chemotherapy (n = 1051) or AC for breast cancer (n = 889) between 9/2013 and 8/2014 were documented retrospectively in 78 practices and 122 hospitals. The sample size was calculated at a 1.25% incidence of cancers of breast, lung, head/neck, stomach and esophagus. Results: 6233 chemotherapy cycles of pts 1940 were evaluable. In the acute phase, the missing (23.6%) or insufficient (2%) application of NK1-RA were the predominant deviations. In 10.9% of cycles corticosteroids (4.3% no DEX, 5.5% underuse, 1.1% overuse ) and in 10.5% 5-HT3-RA (4.4% no 5HT3, 3.8% underuse 2.3% overuse) were not applied according to GL. In the delayed phase in 39.2% of cycles a 5-HT3-RA was used, not in compliance with the GL. In 34.1% of cycles the NK1-RA and in 17.6% a corticosteroid were not given, other than proposed by the GL. Tab. 1. GL adherence (GLA) according to diagnoses

Cancer

Patients, n

Cycles, n

GL adherence (GLA) acute phase,% of cycles

GL adherence (GLA) delayed phase,% of cycles

Breast

900

3233

60.9

46.5

Lung

601

1830

74.7

33.2

Stomach, esophagus

287

769

49.0

14.8

Head and neck

152

401

66.8

25.9

Disclosure: Hartmut Link: Advisory Role: MSD; Financing of Scientific Research: MSD; Expert Testimony: MSD. Petra Ortner: Financing of Scientific Research: MSD, Riemser Pharma für Öffentlichkeitsarbeit. V630

The somatic burden score: A quantitative tool to evaluate the somatic burden due to chemotherapy-induced adverse events Koehler M.1, Fischer T.1, Kropf S.2, Frommer J.3 University Medical Center Magdeburg, Department of Hematology and Oncology, Magdeburg, Germany, 2University Medical Center Magdeburg, Institute for Biometry and Medical Informatics, Magdeburg, Germany, 3 University Medical Center Magdeburg, Department of Psychosomatic Medicine and Psychotherapy, Magdeburg, Germany 1

Introduction: The somatic burden due to chemotherapy-induced adverse events (SB-CHINAE) represents important information for evaluating tolerability in clinical cancer trials. Although there are several established methods like the Common Terminology Criteria for Adverse Events (CTCAE), heretofore no objective, quantitative measurement exists for the SB-CHINAE. To address this shortcoming, we developed the Somatic Burden Score (SBS-AE) that combines the severity grade and duration of an AE. Purposes of our study were the development and validation of the SBS-AE. Methods: SBS-AE’s calculation was based on the number of days of CTCAE-grades of a particular AE. The target value was the weighted, relative duration of an AE-grade using CTCAE v3.0. We applied the SBS-AE in 64 patients with hematological malignancies and high-dose chemotherapy (HDC). At each clinical encounter, the symptom information from five HDC-associated AEs (fever, diarrhea, vomiting, mucositis, and pain) was completed by a nurse and/or a medical oncologist using a pre-printed form during two inpatient treatment intervals with standard chemotherapy and HDC according to the instructions of the Cancer Therapy Evaluation Program of the National Cancer Institute. The ratio measurement scale of the SBS-AE allows all statistical measures using SBS-AE, as all necessary mathematical operations are defined for it. We calculated an Overall-SBS-HDC, defined as the total SB-CHINAE of HDC. To determine SBS-AE’s criterion and construct validity, three self-rating scales and one clinician rating scale were used. Effect sizes as standard measures of clinical relevance were calculated. This study is registered as part of an observational study (DRKS Main ID: DRKS00003453). Results: The SBS-AE’s criterion validity could be verified both with statistical significance and at least medium-to-large effects (p < 0.05, Cohen’s d > 0.79, f2 > 0.18). The quantitative measured SB-CHINAE was equally associated with subjectively assessed physical health-related quality of life (0.15 ≤ R2 ≤ 0.49), and objectively evaluated toxicities (0.48 ≤ R2 ≤ 0.67), transfusion-dependent thrombocytopenia and anemia (Cohen’s d > 0.89). Patients’ somatic burden of HDC was 5.8 fold greater compared with standard chemotherapy regimens. Conclusions: The SBS-AE indicates psychometric and clinical properties and may prove useful in the future design of cancer clinical trials and supportive care interventions inside of the inpatient setting. Disclosure: No conflict of interest disclosed.

In all chemotherapy cycles GLA was significantly higher (p < 0.001) in certified or comprehensive cancer centers (CENT) (71.8%) compared to other institutions (59.3%) in acute phase (OR 1.75; 95% CI 1.57–1.96). In the delayed phase the difference between CENT (45.8%) and other entities (32.5%) was significant (p < 0.001) as well (OR 1.76; 95% CI 1.58–1.96). GLA in the acute phase was significantly lower (p < 0.001) among hematologists/oncologists (58.7%) compared to other medical specialties (69.0%), OR 0.64 (CI 0.58–0.71). In the delayed phase the GLA of hematologists/oncologists was 24.3% compared to others with 47.8% (p < 0.001), OR 0.35 (CI 0.31–0.39). Conclusions: The adherence to current antiemetic guidelines in HEC is different between cancers and medical specialists. The adherence to GL is not sufficient, especially in the delayed phase of HEC.

192

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag

Tumor-/Zellbiologie II V631

The number of tumorspheres cultured from CETCs depends on stage of disease and obtaining chemotherapy in breast cancer patients Pizon M.1, Zimon D.1, Pachmann U.2, Pachmann K.2 SIMFO GmbH, Bayreuth, Germany, 2Transfusionsmedizinisches Zentrum Bayreuth, Bayreuth, Germany 1

Background: Breast cancer is one of the leading causes of cancer death for women worldwide. Major hurdles for a successful treatment are cancer metastasis, resistance to therapy and disease recurrence. The presence of CETCs is closely related to metastasis formation, but the mechanisms through which CETCs promote recurrence of disease are still unclear. Therefore, the aim of this study was to determine the proliferative capacity of CETCs by analyzing the frequency of tumorsphere formation with subsequent phenotypic characterization of the spheres arising in breast cancer patients. Methods: CETCs were cultured under conditions favoring growth of tumorspheres from 72 patients with breast cancer, including a subpopulation of 23 patients with metastatic disease. Cell viability, stem cell marker expression and ALDH 1 activity was evaluated by fluorescence scanning microscope (Olympus Scan®R). Results: Sphere formation was observed in 79% of patients with breast cancer. In the current study we found that the number of tumorspheres depended on stage of disease. Patients in stage IV had statistically significant more tumorspheres compared to patients in stage I (median 6 vs. 2; p = 0.002). The most important factor for growing of tumorspheres is obtaining chemotherapy. Patients with chemotherapy treatment had lower numbers of tumorspheres compared to patients without chemotherapy (median 2 vs. 5; p = 0.002). Interestingly, patients with HER2 positive primary tumor had higher number of tumorspheres with median 10. Analysis of surface marker expression profile of tumorspheres showed that spheres cultured from CETCs had typical phenotype of cancer stem cells with a high enzymatic activity for ALDH 1. There was no sphere formation in a control group with 50 healthy donors. Conclusion: This study demonstrates that a small fraction of CETCs has proliferative activity. Identifying the CETC subset with cancer stem cell properties may provide more clinically useful prognostic information. Chemotherapy is the most important component in cancer therapy because it frequently reduces the number of tumorspheres. Disclosure: No conflict of interest disclosed. V632

Non-malignant MSCs and monocytes support the growth of AML cells in-vitro and in-vivo and Gfi1 regulates the polarization of stromal elements towards a leukemia supporting state Al-Matary Y.S.1, Botezatu L.1, Opalka B.1, Hönes J.1, Thivakaran A.1, Lams R.F.1, Köster R.1, Michel L.C.1, Dührsen U.1, Schroeder T.2, Dürig J.1, Haas R.2, Khandanpour C.1 University Hospital Essen, Hematology, Essen, Germany, 2Heinrich Heine University Duesseldorf/ University Hospital, Clinic for Hematology, Oncology, and Clinical Immunology, Duesseldorf, Germany 1

The growth of solid tumors, lymphoma and leukemias is not only the result of cell-intrinsic genetic and epigenetic alterations, but is also affected by the surrounding stroma and the cells therein. Growth Factor Independence 1 (GFI1), a transcriptional repressor, is involved in differentiation and maturation of various hematopoietic cells, immune cells and mesenchymal stromal cell (MSC). We investigated the role of MSC and monocytes with regard to support of human and murine acute myeloid leukemia (AML) and the role of Gfi1. Bone marrow (BM)-derived

Abstracts

MSCs and monocytes from AML patients better supported growth of a human leukemia cell line in vitro than cells from healthy persons. MSCs and monocytes from patients in remission did not support the growth of leukemia cells to the same extent. To better understand the mechanism behind this, we used different murine MDS and AML models. BM- derived MSC and macrophages from AML mice better supported growth of leukemic cells in vitro than macrophages and MSC from healthy mice. Numbers of monocytes in the BM correlated with aggressiveness of the leukemia. Within the monocyte fraction, the percentage of tumor-associated macrophage (TAM; MHC II- Ly6c-) was significantly higher in the BM and spleen of leukemic than in BM and spleen of healthy mice (p = 0.003). Gfi1 was expressed 3–4 fold higher in the MSC and monocytes from leukemic mice compared to healthy mice. To investigate the role of Gfi1 in the polarization of stroma cells, we transplanted Gfi1-deficient and Wt mice with MLL-AF9 transduced BM cells. We observed a lower number of non-malignant monocytes in the BM of Gfi1-deficient leukemic mice compared to Gfi1 Wt mice pointing to a functional role of Gfi1 in monocyte polarization. Furthermore, Gfi1-deficient mice survived longer than Wt mice. In addition, Gfi1-deficient MSC and monocytes from leukemic mice did not support the growth of leukemic cells in vitro to the same extent as Wt leukemic monocytes. Furthermore, the TAM percentage in BM and spleen of transplanted leukemic mice treated with SH-2251, a compound that dramatically decreases Gfi1 mRNA expression, was lower than in untreated leukemic mice. In summary, MSC and monocytes play an important role in the support of leukemic cells. Gfi1 is involved in the polarization of these cells, and this could serve as a new and additional approach to treat AML patients by impairing the function of leukemia-supporting MSCs and monocytes. Disclosure: No conflict of interest disclosed. V633

Phenotypic assays using zebrafish hematopoesis models for elucidation of hematopoietic toxicity Lenard A.1, McGinnis C.2, Lengerke C.1 Universitätspital Basel, Department of Biomedicine, Basel, Switzerland, 2Roche Innovation Center Basel, Pharmaceutical Sciences DDS-Mechanistic Safety, Basel, Switzerland 1

Introduction: Hematologic findings are one of the most common side effects encountered in preclinical safety testing of new drug candidates. The consequences of direct or indirect damage to blood cells and their precursors can be potentially life-threatening, and hence, hematotoxicity can lead to the termination of a promising drug candidate. Current hematotoxicity testing employs in vitro models with a cell viability read-out. However, this approach only allows a limited read-out and, for example, does not capture effects on later maturation stages of blood cell progenitors. Here, we propose zebrafish as an alternative animal model that captures the full range of hematopoietic lineages and maturation stages in an in vivo setting. Methods: Using flow cytometry and high-content in vivo imaging of fluorescent reporter expression we analyze defined hematopoietic lineages in regards to cell numbers, distribution and activity. Results: Currently, the first set of reference compounds is being evaluated, in order to validate the utility and sensitivity of this model system. A double transgenic reporter system was established for flow cytometry analyses of erythroid lineage (Tg(gata1:DsRed);(globin:GFP)) allowing distinguishment of mature globin/gata1 double positive erythrocytes from gata1-only positive erythroid progenitors. The line was validated in a proof-of-principle phenylhydrazine-induced hemolytic anemia assay where depletion of mature erythrocytes was documented. Furthermore, treatments with antimitotic cancer drugs showed time-dependent depletion of progenitors and/or mature erythroid cells. Additionally, an automated image analysis tool was established for assessment of neutrophil and macrophage numbers using 3D-imaged, live embryos from transgenic lines Tg(mpo:GFP), Tg(mpeg1:Gal4;UAS:Kaede). Further transgenic

Oncol Res Treat 2015;38(suppl 5):1–270

193


CONTENTS AUTHOR INDEX

lines, e.g. for lymphoid and stem cell lineages, are currently generated and assessed for imaging and flow cytometry assays. Conclusions: It is expected that upon successful validation, this new multiplexed in vivo model will find immediate application in predictive and mechanistic preclinical safety testing, complementing existing in vitro hematopoietic approaches and more complex in vivo studies in mammals. Disclosure: No conflict of interest disclosed. V634

Patients with selective IgM deficiency display quantitatively altered B-cell subsets and functional B-cell deficits Mensen A.1, Krause T.1, Hanitsch L.G.1, Meisel C.2, Volk H.-D.1,3, Scheibenbogen C.1,3, Na I.-K.1,4,5 Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Institut für Medizinische Immunologie, Berlin, Germany, 2Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Labor Berlin GmbH, Abteilung Immunologie, Berlin, Germany, 3Berlin Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany, 4Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Abteilung für Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany, 5Charité Universitätsmedizin Berlin, Campus Buch, Experimental and Clinical Research Center (ECRC), Berlin, Germany 1

Introduction: Selective IgM deficiency represents a primary immunodeficiency (sIgM), which is characterized by isolated low IgM level in serum, increased susceptibility to infections and autoimmunity. Although there is some evidence for defects in lymphocyte function, little is known about lymphocyte subset counts and B-cell subset differentiation after T-cell dependent activation. Methods: We determined absolute B and T cells and subset numbers in peripheral blood from 12 adult sIgM patients by flow cytometry. Moreover, flow cytometry and ELISpot analysis were used to assess the differentiation and antibody-secreting capacity of B-cell subsets after seven days in vitro stimulation of PBMCs with CpG, SAC (Staphylococcus aureus Cowan I), PWM (Pokeweed mitogen), IL-2, IL-10, IL-21 and CD40L. Results: All 12 patients had repeatedly diminished serum IgM of ≤40 mg/ dl and normal IgG and IgA levels. We found significantly increased transitional B cells in sIgM patients when compared to numbers of healthy controls (median cells/µl: controls 3.3, sIgM 6.2; p ≤ 0.05). Some patients had decreased numbers either of IgM-expressing switched memory (n = 5), total switched memory (n = 3), or non-switched memory B cells (n = 1). Moreover, memory B-cell subsets expressed significantly lower mIgM levels (MFI), while percentages of mIgM-expressing cells were normal. No significant differences were seen in numbers of naïve, central and effector memory as well as terminal differentiated CD4 and CD8 T-cell subsets. Following in vitro stimulation strong B-cell expansion was seen for healthy controls, whereas no B-cell expansion was observed for 5 of 6 investigated patients after stimulation (median fold expansion: controls 6.7, sIgM 1.4). This diminished expansion was correlated with significantly decreased mIgM-expressing memory B cells and antibody-secreting cells. Two patients showed a severely reduced IgM-secreting capacity as determined by ELISpot assay. Conclusions: Quantitatively altered B-cell subsets, reduced mIgM expression levels and profound deficits of B-cell subsets in the capacity to expand and differentiate into IgM-secreting cells are found in sIgM patients. However, the defects are heterogeneous among the group. These characteristics may provide a basis for diagnosis and classification of sIgM patients to study genetic defects. Disclosure: No conflict of interest disclosed.

194

Oncol Res Treat 2015;38(suppl 5):1–270

V635

Azathioprine-induced severe bone marrow aplasia due to deficiency of thiopurine S-methyltransferase activity Adamek J.1, Schaich M.1, Parmentier S.1 Rems-Murr-Klinik Winnenden, Abteilung für Hämatologie, Onkologie und Palliativmedizin, Winnenden, Germany 1

Introduction: Azathioprine (AZA) – a purine analogue – is commonly used as an immunosuppressive drug in organ transplantation and autoimmune diseases. In vivo, Azathoprine converts rapidly into the pharmacologically active 6-mercaptopurine. The further metabolism of 6-mercaptopurine is complex and depends on different enzymatic pathways. One of these enzymes is the thiopurine S-methyltransferase (TPMT). Due to interindividual variability in TPMT enzyme activity caused by genetic polymorphism there is a diverse receptivity for severe cytotoxic effects. There is a graduation in very low / undetectable, intermediate and high TPMT activity. Patients who are homozygous for the low activity TPMT allele are at high risk to develop severe myelotoxicity because of elevated concentrations of the cytotoxic metabolites. Case description: A 60-year old patient was presented to us with pancytopenia (haemoglobin 7.1 g/dl, platelets 17 tsd/µl, neutrophile granulocytes 0.08 × 103/µl). AZA was started 8 weeks ago because of a clinical aggravation of her Morbus Crohn. The patient reported of increasing fatigue and exertional dyspnoea during the last two weeks. Clinical examination showed petechiae mainly in the lower extremities. A bone marrow aspirate and biopsy after a couple of days without regeneration of the peripheral blood even with G-CSF revealed severe bone marrow aplasia. Responsible for this was a homozygous defect of the TPMT (activity < 5 nmol/g Hb*h). We continued the application of G-CSF. In addition, beside reverse isolation supportive care was applied by prevention of mucositis, application of antifuncal and antiviral prophylaxis and, if necessary, irradiated platelet and red blood cells concentrates were transfused. In the further course, the patient required broad-spectrum antibiotics because of neutropenic fever, but fortunately she developed no severe infections. After three weeks of G-CSF the neutrophile granulocytes showed an adequate increase. Conclusion: In this case, very severe pancytopenia following the treatment with Azathioprine was due to severe deficiency of TPMT activity. This life-threatening complication is rare but predictable and might justify screening patients for TPMT deficiency before starting with AZA in addition to regular routine blood counts when having started a treatment with Azathioprine or 6-Mercaptopurine. Disclosure: No conflict of interest disclosed. V636

Impact of sleep on monocytes Hahn J.1, Günter M.1, Autenrieth S.E.1 Medizinische Klinik / Universität Tübingen, Innere Medizin II, Tübingen, Germany 1

Recent studies have shown that sleep has a strong impact on different components of the immune system. Sleep deprivation has been described to affect both humoral as well as cellular immunity. Previously, the NLR ligand muramyldipeptide (MDP), chemically unique cell wall component of all bacteria, has been identified as a sleep-promoting factor in humans and animals, which accumulates during the active period to eventually induce slow wave sleep (SWS). On the other hand, sleep deprivation leads to a lethal sepsis in rats. Here we addressed whether sleep has an impact on the cellular components in blood and spleen and on the outcome of infection. Our results show that sleep deprivation (SD) in mice for 6 hours led to a significant reduction of cell numbers in the blood in particular of monocytes whereas no effect was seen for PMNs. Further, we observed a reduction of monocytes in the spleen. After intravenous infection of mice with the model pathogen Yersinia enterocolitica, to mimic sepsis, the bacterial load in the spleen of SD-mice was significantly higher one and three days post infection. Also the survival of the SD-mice was signifi-

Abstracts


CONTENTS AUTHOR INDEX

cantly reduced compared to the control group. These data suggest that sleep regulates homeostasis and function of innate immune cells, which are important in the early defence against pathogens. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium AML – Neue Substanzen V637

Sorafenib in AML treatment Röllig C.1, Studienallianz Leukämie (SAL) Universitätsklinikum Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany 1

Introduction: Sorafenib is a multi-kinase inhibitor with activity against several oncogenic kinases that may play a role in the pathogenesis of acute myeloid leukemia (AML). In-vitro data suggest that sorafenib might be an effective drug for the treatment of AML. Since 2009, clinical experience with sorafenib in AML has been reported. Methods: In May 2015, a systematic review based on search for the search terms sorafenib and acute myeloid leukemia in PubMed and Clinicaltrials.gov was conducted. Search results were selected by title and abstract and grouped by study type and corresponding level of evidence. Results: The systematic search delivered 137 publications and 23 registered trials. Of 137 publications, 93 contained reviews or preclinical data, 6 publications were not available in English and 38 reported clinical data. The 38 clinical publications comprised 7 clinical trials and 31 case reports or case series (24 for relapse and 7 for primary treatment). One randomized phase II trial of elderly AML patients has been fully published and another randomized phase II trial in younger patients is available as abstract, both in primary treatment. In these randomized trials by the SAL study group, sorafenib caused a significant prolongation of eventfree survival and relapse-free survival (RFS) in younger AML patients, but showed no beneficial effects and increased toxicity in the elderly trial. Significant RFS improvement in younger patients did not translate into overall survival prolongation. Interestingly, antileukemic efficacy was not restricted to FLT3-ITD mutated AML, suggesting alternative pathways of action. According to the database search, of 23 registered trials testing sorafenib in AML, 10 were still recruiting by May 2015. Conclusions: Sorafenib is efficacious in AML. Moreover, it is the first kinase inhibitor with randomized proof of efficacy in AML. However, not all patients benefit from treatment, and so far trials have failed to show an overall survival benefit. The presented body of evidence supports sorafenib use in patients with relapsed and refractory AML, possibly as a bridging strategy to allogeneic transplantation. The data do not provide a rational for sorafenib as regular component of routine first-line treatment. The future role of the drug in AML treatment will depend on the results of ongoing trials, potential combination partners and the identification of patient subgroups that will most likely benefit from sorafenib.

differentiation. Gemutuzumab ozogamicin (GO) is a combination of the chemotherapy agent calicheamicin and the recombinant humanized IgG4 antibody directed against CD33. The toxicity of calcheamicin is more than 1000 fold higher than that compared to doxorubicin. Thus, the selectively of CD33 to AML blasts is an important requirement for the usage of calcheamicin to avoid overt toxicities. The US Food and Drug Administration (FDA) approved GO for relapsed elderly patients with CD33+ AML at a dose of 9 mg/m2 per day on day 1 and 14 in 2000 based on 3 trials that all used GO as a single agent and showed an increased CR rate. However, the confirmatory trial (SWOG S0106) which was essential for the accelerated approval, showed in the interim analysis no benefit but patients treated with GO also showed an increased early mortality during induction therapy. Based on this analysis, the drug was withdrawn from the market in 2010. Since then, several important trials have given new insights about its utility and toxicity in different settings of AML treatment. From these studies, it can be concluded that GO can lead to an improved outcome in AML patients undergoing intensive therapy only when given in parallel to induction therapy while the positive effect is not seen when given before starting therapy or during consolidation. The dose of GO is important and should be less than 6 mg/m2 as a dose of ≥6 mg/m2 leads to increased toxicity, especially sinusoidal obstructive syndrome (SOS) of the liver, without additional benefit. Furthermore, patients with favorable- or intermediate-risk cytogenetics are more likely to benefit from the addition of GO compared to patients with adverse cytogenetics. Importantly, response to GO does not correlated with CD33 expression and CD33 expression should not guide therapy. Thus, GO is a promising agent for selected patients but as it is currently not approved in Europe it is not easily available outside trials. Disclosure: No conflict of interest disclosed.

Fortbildung

Intensivmedizin V641

Hematological patients on ICU: Developments on the example of center Dresden University Hospital Kroschinsky F.1, Stölzel F.1, Friedrich A.1, Weise M.1, von Bonin S.1, Bornhäuser M.1, Ehninger G.1 Uniklinik Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany

1

Antibody based chemotherapy is becoming an increasingly interesting approach in the field of oncology. Its attractiveness is also due to the fact, that that the antibody allows the delivery of the chemotherapy agent directly and selectively to the target cells. In acute myeloid leukemia (AML), the adhesion protein CD33 is a promising target as it is expressed by approximately 90% of leukemic blasts while its expression decreases with myeloid

Introduction: In 2002 we reported outcome and prognostic features of hematological cancer patients requiring intensive care unit (ICU) treatment between 1995 and 2000 (Kroschinsky et al., Intensive Care Med 2002; 28: 1294–1300). Strategies in cancer treatment, supportive and intensive care as well as patient populations have changed since then. The impact of these developments on patient’s characteristics, type and severity of disorders and outcomes should be studied. Methods: We retrospectively analyzed data of 302 patients with hematological malignancies (including autologous but not allogeneic transplant recipients) undergoing ICU treatment between 01/2001 and 02/2008. The patients’ charts were studied with respect to demographic data, underlying disease, cause of admission, organ dysfunctions, need for mechanical ventilation (MV) and hemofiltration (HF), as well as ICU survival and survival after discharge. Data of these patients (cohort B) were compared to those of our previous study (cohort A, n = 104). Results: Median age in cohort B was 65 (range 19–88) years compared to 55 (range 19–77) years in cohort A. The majority of patients suffered from acute leukemia, aggressive lymphoma or multiple myeloma (A: 72%, B: 63%). The proportion of patients with life-threatening complications during high-dose chemotherapy or who had previous autologous transplantation was 11% in each cohort. Respiratory distress was the main cause of ICU admission in cohort A (48%). Horovitz index < 300 mmHG at ICU admission was documented in 40% of patients in cohort B. Invasive

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: Christoph Röllig: Expert Testimony: Förderung der SORAML-Studie durch Bayer Healthcare Germany. V638

Mylotarg Thol F.1 Medizinische Hochschule Hannover, Hannover, Germany

1

195


CONTENTS AUTHOR INDEX

mechanical ventilation was needed in 52% of cohort A patients vs. 40% of patients in cohort B. In addition, a substantial number of patients of cohort B (148/304, 49%) could be managed with only non-invasive ventilation. Fifty-eight out of 104 (56%) patients in cohort A survived ICU vs. 201/304 (67%) in cohort B. Overall survival at 6 month and 1 year after ICU admission was 33% and 29% in cohort A, and 31% and 22% in cohort B, respectively. Conclusions: Although patients with hematological malignancies requiring ICU treatment were older in the recent analysis, a higher proportion of them survived a life-threatening disorder compared to our previous report. However, long-term outcome after discharge from ICU and hospital did not improve. Further studies are needed to investigate how to overcome the negative impact of ICU treatment on survival. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium MDS Zellregulation V644

The role of the bone marrow microenvironment in the pathogenesis of MDS Schroeder T.1, Geyh S.1, Haas R.1, Germing U.1 Universitätsklinik Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany 1

Myelodysplastic syndromes (MDS) are a heterogenous group of clonal bone marrow disorders characterized by hematopoietic insufficiency and an increased propensity to progress towards AML. While it is a well-accepted concept that MDS originate from hematopoietic stem and progenitor cells (HSPC), the contribution of bone marrow (BM) microenvironment to the pathogenesis of MDS has recently gained more interest. On the one hand, this is based on a better understanding of its physiological role in the regulation of hematopoiesis. Furthermore, it was demonstrated as a ‘proof of principle’ that genetic disruption of cells of the mesenchymal or osteoblastic lineage can induce MDS, MPS or AML in mice. Most work in this field focused on mesenchymal stromal cells (MSC) which are an integral part of this BM microenvironment. Through their spatial proximity within the stem cell niches they bi-directionally interact with HPSC and thereby physiologically influence the behavior of HPSC. Initial investigations had created controversies regarding the contribution of MSC to human MDS pathogenesis. Still, we and other have recently demonstrated that MDS-derived MSC are structurally, molecularly and functionally altered. As a consequence, MDS-derived MSC provide insufficient stromal support for healthy HSPC thereby substantially contributing to insufficient hematopoiesis, which is the hallmark of MDS. In addition to the insufficient support of normal hematopoiesis, sophisticated experiments nicely showed that the interaction between hematopoietic and stromal cells is needed to ensure engraftment of clonal MDS cells in mice. These novel insights have raised the important question whether MSC carry primary defects contributing to hematopoietic failure in MDS, or whether the observed alterations represent secondary adaptations to the expanding MDS clone. Experiments to answer this question are currently running and their results will hopefully contribute to the development of novel treatment approaches which aim to abrogate the self-reinforcing MDS BM microenvironment and to restore normal hematopoiesis. Disclosure: Thomas Schroeder: Advisory Role: Celgene, Novartis; Financing of Scientific Research: Celgene; Expert Testimony: Celgene; Other Financial Relationships: Celgene Reiseunterstützung. Ulrich Germing: No conflict of interest disclosed.

196

Oncol Res Treat 2015;38(suppl 5):1–270

Wissenschaftliches Symposium Supportive Therapien V649

Hemoglobin transfusion triggers in hematology and oncology Schuler U.S.1 Universitätsklinikum Carl Gustav Carus, Dresden, Medizinische Klinik I und PalliativCentrum, Dresden, Germany 1

In recent years, several studies have addressed the impact of transfusion triggers for thrombocytes in Hem/Onc, but there is a paucity of data concerning triggers for erythrocyte concentrates. Historically in most medical contexts triggers as high as 10 g/dl were used, but RCTs comparing such liberal with more restrictive triggers have frequently shown advantages for the more restrictive strategy. An increased rate of infection, as a consequence of immunomodulation by transfusions seems to play a role. It is unclear, whether this effect is of importance in already highly immunosuppressed patients with hematological malignancies or intensive chemotherapies for solid tumours. Based largely on data from other clinical settings several guidelines have advocated triggers as low as 7 g/dl (4,34 mmol/l) and even lower in acute situations. For example the cross-sectional guideline (1) of the German Medical Association (Querschnitts-Leitlinie BÄK zur Therapie mit Blutkomponenten und Plasmaderivaten) sees the decision largely dependent on the patient’s capacity to compensate anemia and on patient risk factors. According to the guideline the only definite trigger would be < 6 g/dl (< 3,7 mmol/l). With higher hemoglobin levels the patient’s capacity for compensation and risk factors become essential. Compensation is judged by factors like cardiopulmonary symptoms [tachycardia, hypotension, dyspnea], ECG-changes [ST-segment elevations and depression, arrhythmia], in ICU-environments more sophisticated parameters come into play (Oxygen extraction ratio and others). In situations of chronic anemia, transfusions are recommended for a range of triggers, Hk < 24–21% or Hb < 8–7g/dl (<5,0–4,34 mmol/l). For patients with cardiovascular diseases, one should keep in mind that RCTs so far have shown the safety of restrictive approaches down to 8 mg/dl (2) only. In hematology for example RCTs in leukemia therapy comparing meaningful triggers of 7 g/dl versus 8–9 g/dl are desirable. In SCT a very small RCT has associated an extremely high trigger of 12 g/dl with an increased incidence of VOD (3). Survival outcomes, infection rates, patient reported outcomes (QOL) and indices of iron overload should be monitored. References: 1 w ww.bundesaerztekammer.de/fileadmin/user_upload/downloads/ QLL_Haemotherapie_2014.pdf. 2 Carson et al.: Lancet. 385(9974);2015:1183. 3 Robitaille et al.: Biol Blood Marrow Transplant. 19(3);2013:468–473. Disclosure: No conflict of interest disclosed. V650

Biosimilars and originator biologicals- equivalence in anemia and neutropenia? Link H.1 Westpfalz-Klinikum Kaiserslautern, Medizinische Klinik I, Kaiserslautern, Germany 1

Biosimilars are similar biological medicinal products submitted to a specific approval pathway in the EU and the USA that are demonstrated to be “biosimilar” to or interchangeable” with a licensed biological product. Biologicals are derived from living cells or organisms and consist of relatively large and highly complex molecular entities that are often difficult to fully characterize. The inherent variability of the biological system used as manufacturing process, results in a biological product that will also vary to a certain degree (´microheterogeneity´). The European Medicines Agency (EMA) guideline for the development of biosimilars requires numerous functional and clinical studies. Of note, a comparability exercise is

Abstracts


CONTENTS AUTHOR INDEX

also required for originator biologicals when changes to the manufacturing process are made, such changes are frequently introduced throughout a biological’s lifecycle. If clinical similarity can be shown in a key indication, extrapolation of efficacy and safety data to other indications of the reference product may be possible. Clinicians mainly consider the clinical trial data to judge the efficacy and safety of a medicinal product. Recombinant erythropoietins (epoetins) heavily glycosylated and rather complex molecules. EMA guideline for the development of biosimilar epoetins requires clinical studies, including at least one clinical trial in patients with renal anemia who do not have major complications that may impair the response to epoetin. There are rational scientific arguments substantiating the extrapolation from the renal anemia to the cancer indication There are no specific data concerning efficacy or safety issues clinical practice for biosimilar epoetins licensed in Europe. Filgrastim is approved for treatment of neutropenia after chemotherapy and for the mobilization of peripheral blood progenitor cells in patients and healthy donors. Several biosimilar filgrastims have been licensed in the EU and one in the USA, and in all cases, all indications of the originator product were approved. Post marketing studies confirmed efficacy and safety of biosimilar filgrastim products in the approved indications, including the mobilization of stem cells in healthy donors. In summary biosimilar epoetins and figrastims approved by the EU or FDA are efficacious and safe products.

months (range: 0.4 to 192 months), 24% of patients had died. The 1-year overall survival was 87%, after 2 years 79% and after 3 years 76% of patients were still alive. The median survival for patients with relapse was 22 months (95% CI: 13–32 months). The 2-year survival for patients with relapse was 33% and was significantly lower than the 2-year survival of 90% for patients without relapse. Significant prognostic markers for OS were B-symptoms (univariate [uv] p = 0.012), grade of remission (uv p < 0.001), occurrence of relapse (uv p < 0.001) and LDH concentration (uv, metric: p = 0.003) were identified. Results of the multivariate Cox regression analysis showed that LDH concentration at initial diagnosis of ML (multivariate p = 0.046) and relapse (multivariate p = 0.050) as independent factors in respect to survival. Patients treated in the Univ. of Heidelberg and the Mannheimer Onkologie Praxis had an identical prognosis. Conclusions: First-line treatment of ML in HIV-positive patients is of critical importance. Patients who achieved and maintained a first CR had an excellent prognosis.

References: Weise, M., et al.: Blood, 120 (26);2012:5111–5117. Weise, M., et al.: Blood, 124 (22);2014:3191–3196.

Liposomal formulation of vincristine allows for doubling the dose compared to conventional vincristine: Results of the first futility analysis of the OPTIMAL>60 study of the German HighGrade Non-Hodgkin Lymphoma Study Group (DSHNHL)

Disclosure: Hartmut Link: Advisory Role: Amgen, Hexal (Sandoz), Teva; Financing of Scientific Research: Amgen, Hexal (Sandoz), Teva; Expert Testimony: Amgen, Hexal (Sandoz), Teva.

Freier Vortrag

Lymphome aggressiv klinisch II V655

Excellent prognosis of patients with malignant lymphoma and HIV infection reaching a first remission – a retrospective data analysis of 50 patients from two centers Buss E.C.1, Hering J.1, Ho A.D.1, Hensel M.2, Witzens-Harig M.1 Universitätsklinikum Heidelberg, Medizinische Klinik V, Heidelberg, Germany, Mannheimer Onkologie Praxis, Mannheim, Germany

1 2

Introduction: Patients with HIV infection are significantly more likely to suffer from newly diagnosed malignant lymphoma (ML) than other individuals and ML is still the leading cause of death among AIDS related diseases. Methods and Results: The work presented here is based on data from 50 HIV-positive patients with various types of ML. Clinical data was retrospectively collected of patients of the University Hospital Heidelberg and from the Mannheimer Onkologie Praxis. Male gender prevailed with 86%. Median age at diagnosis of ML was 43.5 years with a median interval between the diagnosis of HIV and ML of 4 years (range: 0–29 years). 28% of patients were diagnosed with HIV in the context of ML diagnosis. 16% of patients were diagnosed with Hodgkin´s lymphoma [HL], 42% DLBCL, 26% Burkitt´s lymphoma, 12% plasmoblastic lymphoma, 5% primary lymphoma of the cerebral nervous system, 2% MM, 2% MALT and 2% FL. The ORR of patients analyzed within this study was 91%. 24/43 patients achieved a CR after treatment and 15/43 patients a PR (56% and 35%). 7% of patients suffered from PD. Relapse occurred in 18% of patients, all male, and in 65% of patients with PR (5 years cumulated). Treatment was with standard protocols, in 72% of patients with concurrent antiretroviral therapy (cART), 9% without concurrent cART and 16% unclarified status in respect to cART. After a median follow-up of 31

Abstracts

Disclosure: Eike Buss: Immaterial Conflict of Interests: Mitglied GMALL ­Protokollkommission, lokaler PI GMALL. Mathias Witzens-Harig: Financing of Scientific Research: Roche; Immaterial Conflict of Interests: PI mehrerer Lymphomstudien. V656

Duecker S.1, Poeschel V.1, Wolf A.1, Held G.1, Murawski N.1, Zwick C.1, Haenel M.2, Viardot A.3, Truemper L.4, Ziepert M.5, Pfreundschuh M.1, Deutsche Studiengruppe Hochmaligne Non-Hodgkin Lymphome (DSHNHL) Klinik für Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany, 2Klinik für Innere Medizin III, Klinikum Chemnitz, Chemnitz, Germany, 3 Abteilung für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany, 4 Abteilung Hämatologie und Onkologie, Universitätsmedizin Göttingen, Göttingen, Germany, 5Institut für Medizinische Informatik, Statistik und Epidemiologie (IMISE), Leipzig, Germany 1

Background: Cumulative neurotoxicity is a dose limiting factor of vincristine, especially in elderly patients. In the RICOVER-60 trial of the DSHNHL [Pfreundschuh et al.: Lancet Oncol. 2008] 22% of patients received no vincristine at cycle 6 and 36% at cycle 8 due to neurotoxicity. On the other hand, retrospective analyses suggest that dose reductions of vincristine lead to a considerable loss of efficacy. Since preclinical and clinical studies suggested high activity and excellent tolerability of liposomale vincristine (VLIP), a randomized comparison of the two formulations was warranted. Methods: In the OPTIMAL>60 study elderly patients with DLBCL (61to 80 years-old, ECOG 0–4, IPI 1 [age > 60] with bulk, or IPI 2–5) are randomized to eight 2-week applications or an optimized schedule of 12 applications of rituximab in combination with 6 cycles CHOP-14. Patients receive a second randomization into conventional vincristine (VCR, 1.4 mg/m2, capped at 2 mg) or VLIP (2 mg/m2, uncapped). The determination of the maximum median doses VLIP and VCR is a secondary endpoint. Neurotoxicity was determined as polyneuropathy according to the Common Terminology Criteria for Adverse Events (CTCAE v. 4.03). Results: In this planned futility analysis fifty-three patients received VCR and 52 patients VLIP. The median cumulative dose of VLIP (10.09 mg/m2) was nearly double the dose of the VCR (5.42 mg/m2) (p < 0.001). Since identical dose reduction rules were applied, neurotoxic effects with VLIP and VCR were identical. After cycles 1 and 2 the median grade of polyneuropathy was 0 in both groups, which increased to a median toxicity of grade 1 after cycles 3, 4, 5 and 6. The maximum grades of polyneuropathy observed in both groups were not significantly different (p = 0.865). No VLIP was administered in 18 patients (34.62%), no VCR in 14 patients (26.42%) in at least one cycle of therapy (p = 0.361) due to polyneuropathy.

Oncol Res Treat 2015;38(suppl 5):1–270

197


CONTENTS AUTHOR INDEX

Conclusions: With nearly twice the dose of VLIP achieved in elderly patients treated in the OPTIMAL>60 study, the preset goals were met and the study continues as planned. A total recruitment of 864 patients is necessary to demonstrate whether the double dose of VLIP will translate into a significantly improved outcome in 3-year PFS (HR 0.68 or 9% with an alpha = 5% and a power of 80%). Acknowledgement: This study was supported by Spectrum, Amgen and Roche. Disclosure: Sascha Duecker: No conflict of interest disclosed. Michael Pfreundschuh: Advisory Role: Amgen, Roche; Financing of Scientific Research: Roche, Amgen; Expert Testimony: Amgen, Roche, Spectrum. V657

Lymphoma-associated mortality in the German HIV-lymphoma cohort study Hentrich M.1, Schommers P.2, Gillor D.2, Müller M.3, Stoehr A.4, Schultze A.5, Jensen B.6, Wasmuth J.-C.7, Wolf T.8, Siehl J.9, Oette M.10, Taylor N.11, Zinngrebe B.12, Hensel M.13, Horst H.-A.14, Fätkenheuer G.2, Wyen C.15, Hoffmann C.16 Rotkreuzklinikum München, III. Medizinische Abteilung, München, Germany, Universitätsklinik Köln, Klinik I für Innere Medizin, Köln, Germany, 3Vivantes Auguste-Viktoria-Klinikum, Abteilung für Gastroenterologie und Infektiologie, Berlin, Germany, 4ifi-Institut für Interdisziplinäre Medizin, Hamburg, Germany, 5 Universitätsklinikum Hamburg-Eppendorf, Klinik für Hämatologie, Onkologie und Knochenmarktransplantation, Hamburg, Germany, 6Universitätsklinik Düsseldorf, Klinik für Gastroenterologie, Hepatologie und Infektiologie, Düsseldorf, Germany, 7Universitätsklinik Bonn, Klinik I für Innere Medizin, Bonn, Germany, 8Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt, Germany, 9Ärzteforum Seestraße, Berlin, Germany, 10Krankenhaus der Augustinerinnen, Klinik für allgemeine Innere Medizin, Gastroenterologie und Infektiologie, Köln, Germany, 11Universitätsklinik Salzburg, Universitätsklinik für Innere Medizin III, Salzburg, Austria, 12Evangelisches Krankenhaus Bielefeld, Klinik für Innere Medizin, Hämatologie/Onkologie und Palliativmedizin, Bielefeld, Germany, 13Mannheimer Onkologie Praxis, Mannheim, Germany, 14 Universitätsklinikum Schleswig-Holstein, Campus Kiel, Klinik für Innere Medizin II, Kiel, Germany, 15Universitätsklinik Köln, Klinik I für Innere Medizin, Kön, Germany, 16IPM Studienzentrum, Infektionsmedizinisches Zentrum Hamburg, Hamburg, Germany 1 2

Introduction: The risk of Non-Hodgkin lymphoma (NHL) and Hodgkin lymphoma (HL) is increased in HIV-infected individuals. The outcome of HIV-associated lymphoma has undergone significant improvement in recent years beginning with the widespread use of combination antiretroviral therapy (ART). However, aggressive lymphomas remain the most frequent AIDS-defining event leading to death. We aimed to analyse lymphoma-related mortality in the German HIV-Lymphoma Cohort Study. Methods: This prospective multicenter cohort study includes adult HIV1 infected patients (pts) with biopsy or cytologically proven HIV-related lymphoma diagnosed at 33 participating centers in Germany and Austria since January 2005. Data on HIV-infection and lymphoma characteristics, treatments and outcomes recorded until December 2014 were recorded. Pts with indolent lymphomas and primary central nervous system lymphomas were excluded from the present analysis. Results: Of 499 pts (463 males, 36 females) 394 had aggressive NHL and 105 HL. At the time of lymphoma diagnosis the median age was 44.7 years (range, 22–74.7) and the median CD4-cell count was 210/µl (0–1586). 344 of 499 pts (69%) were diagnosed with advanced stage (III/IV) lymphoma. 214 pts (43%) were ART-naïve, 175 pts (35%) had a viral load below the detection limit of <50 HIV RNA copies/ml at lymphoma diagnosis while on ART, and 110 pts (22%) had a viral load >50 copies/ml while on ART or while discontinuing ART. After a median follow-up of 1.8 years 25 of 105 HL (24%) and 122 of 394 NHL pts (31%) have died with an overall death rate of 30% (147/499). 84 of 147 pts (57%) died of relapsed/refractory lymphoma, 75 of 122 pts with NHL (62%) and 9 of 25 pts with HL (36%). In 30 of 147 pts (20%) the cause of death were infections during or shortly after primary chemotherapy. Further causes of death were secondary malignancies (n = 6; 4%), AIDS-defining events other than NHL

198

Oncol Res Treat 2015;38(suppl 5):1–270

(n = 6; 4%) including 4 cases of progressive multifocal leukoencephalopathy, and miscellaneous (n = 21; 14%). Conclusions: HIV-related lymphomas mainly occur in ART-naïve or insufficiently treated patients. The major cause of death is relapsed/refractory lymphoma followed by infections during or shortly after primary chemotherapy. Disclosure: No conflict of interest disclosed. V658

Upfront intensification incorporating dose dense rituximab and high dose MTX to CHOP-14 in young high risk DLBCL patients Strüßmann T.1, Fritsch K.1, Fietz T.2, Finke J.1, Marks R.1 Uniklinik Freiburg / Innere Medizin / Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 2Onkologische Gemeinschaftspraxis, Singen, Germany 1

Introduction: Despite treatment of diffuse large B cell lymphoma has improved by incorporating rituximab to CHOP therapy, outcome in young high risk pts remains poor. Earlier studies suggesting superior results using upfront ASCT were hampered by a putative bias in treatment assignment. To overcome this problem we tested a dose-intensified treatment schedule consisting of 6 cycles CHOP-14 with dose dense rituximab as suggested by Pfreundschuh at al. and high dose MTX as CNS directed approach, to be used for all young high risk DLBCL pts Methods: All pts (n = 21) aged < 65 y presenting with high risk DLBCL at Freiburg University Medical Center since 2012 were intented to be treated (ITT) with 6 cycles CHOP-14 and dose dense rituximab (375 mg/m²) on days 0, 1, 4, 8, 15, 22, 29, 47, 61 and 75. HD MTX (3.0 g/m²) was administered on days 30 and 76 right before CHOP. In clinical practice, 3 pts (16%), despite presenting with DLBCL, were not treated according to the protocol. Of those, 1 pt had a double hit lymphoma, and1 pt had a history of prior kidney transplantation. Response was evaluated using PET-CT. Results: Of 18 pts administered to the protocol median age at diagnosis was 52 y with 50% female. Stage I/II vs. III/IV: 22% vs. 78%; B-symptoms: 44%; LDH was elevated in all pts; ECOG > 1: 33%; Extranodal site ≥ 2: 61%; According to IPI and aaIPI 16 pts (89%) had an intermediate high and high risk score. Median follow-up was 15.6 month (0.4–35.9). 16 pts (89%) completed the protocol. No treatment related deaths were observed. Toxicities were as expected and manageable; one pt could not receive 6th cycle CHOP due to severe infection. One early death was observed 4 days after 1st R-CHOP due to lymphoma infiltration of the heart and consecutive arrhythmia. All pts responded, 13 pts achieving a CR (72%) and 4 pts a PR (22%). Of 4 pts in PR by PET criteria 3 pts underwent second biopsy. 2 pts were lymphoma negative and 1 pt positive, resulting in 83% ITT-patients free from disease after induction therapy. The lymphoma positive pt underwent radiation of residual lymphoma masses, achieving a CR. For all pts estimated 18m-OS was 94.5% and 2y-PFS was 94.5%. No CNS events occurred. Conclusions: In routine clinical practice this intensified protocol is feasible for all high risk DLBCL pts < 65 y, showing promising results regarding ORR, PFS and OS. In our cohort no CNS relapse was yet observed. Longer follow-up is required to confirm our results. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V659

Minimal renal toxicity after Rituximab-DHAP with a modified cisplatin application scheme in patients with relapsed and refractory diffuse large B-cell lymphoma Lisenko K.1, McClanahan F.2, Schöning T.3, Schwarzbich M.-A.1, Cremer M.1, Dittrich T.1, Ho A.D.1, Witzens-Harig M.1 Heidelberg University Hospital, Department of Medicine V, Heidelberg, Germany, 2Barts Cancer Institute – a CR-UK Centre of Excellence, Centre for Haemato-Oncology, London, United Kingdom, 3Heidelberg University Hospital, University Pharmacy, Heidelberg, Germany 1

Background: Rituximab in combination with DHAP is a widely accepted salvage regimen in patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL). DHAP combines cisplatin (100 mg/m2) typically administered intravenously (i. v.) by continuous infusion over 24 hours, followed on day 2 by cytarabine (2 g/m2) in a 3-hour infusion repeated after 12 hours, and oral administration of dexamethasone (40 mg/d) for 4 consecutive days. A common adverse effect of this protocol consists of renal toxicity which may result in dose reduction or treatment discontinuation. Therefore novel approaches to overcome renal toxicity of R-DHAP are urgently warranted. Assuming that a lower single dose of cisplatin over several days would reduce renal toxicity, our institution has chosen to administer cisplatin at a dosage of 25 mg/m2 per day as a 3-hour infusion over 4 consecutive days. In this study we systematically examine efficacy and renal toxicity of this modified R-DHAP regimen. Methods: We retrospectively analyzed data of 122 patients with relapsed/ refractory DLBCL who were treated at our institution from July 2002 to July 2013. Patients were grouped according to the number of R-DHAP courses applied and renal function was evaluated in each subgroup. Creatinine serum levels before each R-DHAP cycle and two to three weeks after the last R-DHAP were assessed and GFR was calculated according to the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation. Results: Overall, 256 R-DHAP cycles were administered. 31 (25.4%), 61 (50.0%), 14 (11.5%) and 16 (13.1%) patients received one, two, three or four R-DHAP courses, respectively. Dose adjustments were applied in 5 (4.1%) patients who in total received 11 R-DHAP courses. A step-by-step evaluation of renal function after each R-DHAP course revealed that a GFR decrease can be observed after each chemotherapy cycle. Of note, in none of the subgroups the GFR was lower than 60 ml/min/1.73 m2. In most patients, only renal impairment grade I and II was observed. Renal impairment grade III was observed in 12 patients (9.8%) and grade IV only in one patient (0,8%). The overall response rate of the modified R-DHAP protocol was 54.6% (CR 16.2%, CRu 5.1%, PR 33.3%, SD 15.4% and PD 29.9%). Conclusion: A modified R-DHAP regimen with administration of cisplatin 25 mg/m2 over 4 consecutive days is safe in relapsed/refractory DLBCL and leads only to minimal renal toxicity.

critical to overcome drug insensitivity and may direct the development of novel therapies. Since patient samples are rarely available as matched pairs at diagnosis and at a resistant state, and cannot be further drug-challenged or subjected to functional validation experiments, we considered mouse models as valuable tools for the molecular dissection of treatment responsiveness. We utilize genomics, transcriptomics, proteomics, kinomics and metabolomics in an integrative “pan-omics” approach to decipher mechanisms of treatment resistance in the Eµ-myc transgenic lymphoma mouse model with previously documented cross-species predictability for human diffuse large B-cell lymphomas (DLBCL). Methods: 79 immunocompetent recipient mice were transplanted with 34 primary Eµ-myc B-cell lymphomas, exposed to cyclophosphamide (CTX) upon tumor manifestation, and long-term monitored for response. Whole-exome sequencing, array-based copy number analyses, transcriptomics and kinomics, as well as mass spectrometry-based proteomics and metabolomics were conducted, and the data subjected to bioinformatics processing to unveil mechanisms of treatment resistance. Results: After treatment of lymphoma-bearing mice, lasting remissions (reflecting cure) were observed in about half of them. Repetitive treatment of mice harboring relapse lymphomas resulted in progressively shortened remission times and finally led to full-blown resistance, thereby recapitulating clinical courses of patients with drug-insensitive aggressive lymphomas. Multiple omics technologies were applied to compare curable vs. relapse-prone and resistant lymphomas, all with or without an additional short-term exposure to CTX to acutely challenge drug-specific response programs. First analyses of the data by integrative bioinformatics approaches unveiled candidate mechanisms that currently undergo functional validation. Conclusions: Eµ-myc lymphoma-bearing mice treated in a clinical trial-like fashion were established as a versatile model of clinical chemoresistance. Going beyond a transcriptome-restricted investigation, our pan-omics strategy aims to dissect underlying mechanisms that will be further exploited as (co-)targets for novel lesion-based therapies in future cancer precision medicine. Exemplary pan-omics-derived findings will be presented at the conference. Disclosure: No conflict of interest disclosed.

Freier Vortrag CLL experimentell V661

Neutralization of B cell activating factor (BAFF) by Belimumab sensitizes chronic lymphoid leukemia (CLL) cells to systemic treatment modalities Wild J.1, Strunz B.1, Kousis P.1, Kanz L.2, Salih H.1,2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Department for Internal Medicine II, Eberhard Karls University, Tuebingen, Germany, 2Eberhard Karls University Tuebingen, Department of Hematology and Oncology, Tuebingen, Germany 1

Disclosure: No conflict of interest disclosed. V660

An integrative and functional pan-omics approach to treatment failure in aggressive B-cell lymphomas in vivo

Introduction: Treatment failure is the key determinant of poor outcome in lymphoma therapy. Unveiling the underlying molecular mechanisms is

The TNF family member BAFF contributes to disease pathophysiology of mature B cell malignancies by sustaining survival and preventing apoptosis of the malignant B cells, and elevated BAFF serum levels have been reported in B cell lymphoma patients. Recently, we demonstrated that BAFF protects CLL cells from Rituximab (R)-induced NK cell killing, a major mechanism by which this CD20-antibody mediates its therapeutic effects. Notably, sensitivity of CLL cells to NK cell antibody-dependent cellular cytotoxicity (ADCC) could be restored by the anti-BAFF antibody Belimumab (Benlysta®), which is approved for the treatment of systemic lupus erythematosus (Wild et al, Leukemia 2015). Here we studied whether neutralization of BAFF by Belimumab could also serve to increase the susceptibility of malignant B cells to chemotherapeutic compounds routinely used in CLL therapy. To this end, we exposed CLL cells of leukemia patients ex vivo to chlorambucil and fludarabin/cyclophosphamide (FC)

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Kase J. , Herrmann A. , Fan D.N.-Y. , Lenze D. , Lisec J. , Dittmar G. , Fröhler S.3, Chen W.3, Steinemann D.4, Hummel M.2, Schlegelberger B.4, Leser U.5, Dörken B.1,3, Schmitt C.1,3 1

1

1

2

1

3

Charité – Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumor Immunology and Molekulares Krebsforschungszentrum, Berlin, Germany, 2Charité – Universitätsmedizin Berlin, Institute of Pathology, Department of Molecular Biology, Berlin, Germany, 3Max-Delbrück-Center for Moleculare Medicine, Berlin, Germany, 4Hannover Medical School, Institute of Cell and Molecular Pathology, Hannover, Germany, 5Humboldt Universität zu Berlin, Department of Knowledge Management in Bioinformatics, Berlin, Germany 1

199


CONTENTS AUTHOR INDEX

with and without R in varying concentrations based on plasma peak levels achieved in leukemia patients. Exposure to BAFF protected the primary CLL cells from the effects of the therapeutic compounds on leukemia cell metabolic activity and cell death, and the efficacy of treatment could be restored by Belimumab. Next we employed a NOD SCID IL2Rgc ko (NSG) mouse model to study the effects of BAFF and its neutralization on the survival of transferred patient CLL cells in vivo. In line with our in vitro findings, BAFF protected the leukemic cells from R-FC induced cell death, and Belimumab restored the susceptibility of the CLL cells to systemic treatment. Together, our findings demonstrate that BAFF neutralization can serve to increase the efficacy of systemic treatment modalities used in CLL therapy and warrant future clinical studies of combinatorial approaches which, based on our findings, are presently in preparation. Disclosure: No conflict of interest disclosed. V662

NFAT2 regulates anergy induction in CLL through Lck Märklin M.1, Heitmann J.1, Ganser M.1, Bugl S.1, Kopp H.-G.1, Kanz L.1, Rao A.2, Wirths S.1, Müller M.R.1 Universitätsklinikum Tübingen, Medizinische Klinik und Poliklinik, Dept. of Hematology, Oncology and Immunology, Tübingen, Germany, 2La Jolla Institute of Allergy and Immunology, La Jolla, United States 1

NFAT2 is a highly phosphorylated transcription factor which regulates developmental and activation programs in diverse cell types. CLL constitutes a heterogeneous disease with some patients exhibiting an indolent course for many years and others progressing rapidly and requiring early treatment. A defined subgroup of patients shows enhanced responsiveness to stimulation of the B cell receptor (BCR) complex and more aggressive disease. In contrast, another subset of CLL patients with more indolent course is characterized by an anergic B cell phenotype referring to B cell unresponsiveness to IgM ligation. Here, we analyzed the role of NFAT2 in the pathogenesis of CLL and in anergy induction in CLL cells. We crossed conditional CD19-Cre NFAT2 knockout mice to limit deletion to the B cells lineage with the Eµ-TCL1 transgenic mice, which develop a human-like CLL. We analysed TCL1+NFAT2 ko mice and TCL1 mice without a NFAT2 deletion served as controls. We performed a comparative gene expression analysis and to assess the anergic phenotype in CLL cells and the role of NFAT2 in its induction, we performed Ca2+ mobilization assays using a flow cytometric approach and performed Western Blots for multiple downstream signaling molecules. Mice with NFAT2 ko exhibited a significantly more aggressive disease course with accelerated accumulation of CLL cells and a dramatically reduced life expectancy. We detected a substantially altered expression profile of genes associated with B cell anergy in the TCL1+NFAT2 ko mice. The vast majority of these genes was expressed significantly less in the absence of NFAT2 with Lck, Pacsin1 and Cbl representing the biggest hits. While anergic CLL cells from TCL1 mice exhibited an unresponsive phenotype with respect to Ca2+ flux upon IgM ligation, TCL1+NFAT2 ko mice showed a normal capacity to mobilize Ca2+ and a normal activation of the downstream signaling. IgM stimulation did not activate normal phosphotyrosine induction in TCL1 mice. However TCL1 mice showed a strong activation of the anergy regulator Lck. Genetic loss NFAT2 leads aggressive disease and controls the expression of several important anergy-associated genes. We identified Lck as a critical target of NFAT2 in this context. Taken together, our data demonstrate that the NFAT2-Lck axis plays an essential role in the pathogenesis of CLL and implicate it as a potential target in its treatment. Disclosure: No conflict of interest disclosed.

200

Oncol Res Treat 2015;38(suppl 5):1–270

V663

The conditional loss of Tp53 or Atm in theEµ:Tcl1 mouse leads to the development of an aggressive, chemotherapy-resistant CLL Knittel G.1, Liedgens P.2, Reinhardt H.C.1 Uniklinik, Köln, Germany, 2Universitätsklinikum Köln, Medizinische Klinik 1, Köln, Germany 1

CLL is the most common leukemia in the Western world. During CLL development, incipient CLL cells undergo a multistep mutational process, during which they acquire a set of genetic and/or epigenetic lesions, which ultimately result in the leukemic state. CLL-associated mutations can be classified into so-called driver mutations, which are essential for malignant growth and passenger mutations, which are functionally less significant. Recent work has revealed the identity of early, so-called trunk lesions and later-occurring subclonal mutations that are associated with disease progression and (chemo-)therapy resistance. Prominent examples for subclonal additional genetic events are ATM and TP53 mutations, which are both associated with resistance against genotoxic therapies. With this genomic information in hand, we now aimed atovercoming one of the biggest hurdles in preclinical CLL research – the lack of mouse models that faithfully mimic the genetic events leading to CLL development. We have generated an characterized CLL models in which the conditional, B cell-specific loss of Atm or Tp53 leads to the development of an aggressive CLL. Furthermore, we can show that specifically the loss of Tp53 leads to the development of Richter syndrome in approx. 20 percent of our animals. Lastly, we use these novel alleles as a preclinical platform for the development and validation of novel targeted therapies. Our novel models now provide a lineup of preclinical tools that mimic both indolent disease, as well as highly aggressive and transformed disease. Disclosure: No conflict of interest disclosed. V664

Integrin-linked kinase is induced by TNFα-NF-κB signals and required for centrosome clustering and mitotic spindle organization during chronic lymphocytic leukemia cell cycle progression Krenn P.1, Hofbauer S.1, Pucher S.1, Hutterer E.1, Hinterseer E.1, Asslaber D.1, Sternberg C.2, Neureiter D.3, Aberger F.2, Wickstroem S.4, Greil R.1, Hartmann T.N.1 Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Paracelsus Medical University, Salzburg, Austria, 2Department of Molecular Biology, Division of Molecular Tumor Biology, University of Salzburg, Salzburg, Austria, 3Institute of Pathology, Paracelsus Medical University, Salzburg, Austria, 4Max Planck Institute for Biology of Ageing, Cologne, Germany 1

Introduction: Integrin-linked kinase (ILK) is a multifunctional intracellular adaptor protein downstream of beta1 integrins and is involved in tumor cell migration and cell cycle progression. The proliferation of chronic lymphocytic leukemia (CLL) cells is promoted by activating signals from immune and stromal cells within the lymphoid microenvironment. Here, we investigated ILK expression, regulation, localization and function during this microenvironmental crosstalk of CLL cells. Methods: ILK expression in peripheral blood (PB) CLL cells was cytometrically assessed. Co-expression of ILK, Ki-67 and the NF-kappaB subunit p65 in CLL patient derived lymph nodes (LNs) was immunohistochemically analyzed. The LN microenvironment was mimicked in vitro by co-cultures of CLL cells with activated T cells or stimulating CLL cells by IL2/CpG. The role of TNFalpha and NF-kappaB was studied using blocking antibodies, specific inhibitors and ChIP assays. Analysis of co-localization of ILK and the centrosomal marker gamma-tubulin was performed by immunofluorescence. ILK function was analyzed in lentiviral transfected Mec1 cells harboring an inducible ILK shRNA construct. Results: We found increased ILK expression in PB CLL cells displaying markers of poor prognosis. Furthermore, CLL cells with a proliferative

Abstracts


CONTENTS AUTHOR INDEX

signature expressed higher ILK than quiescent cells. Consistently, immunohistochemical analysis of CLL patient derived LNs revealed pronounced expression of ILK and the NF-kappaB subunit p65 in regions of increased proliferation. In activating in vitro co-cultures, ILK expression in CLL cells was elevated alongside with Cyclin D1 upregulation. This induction was based on NF-kappaB activation by soluble co-culture produced TNFalpha. A ChIP array validated the direct interaction of p65 and the ILK promoter cis region. The newly synthesized ILK protein co-localized to centrosomal structures required for cell division. Upon an inducible ILK knockdown in Mec1 cells, centrosome clustering was disturbed resulting in disorganized mitotic spindles. Conclusion: In summary, we describe a TNFα-NF-κB mediated induction of ILK during cell cycle progression of CLL cells, which is required for correct centrosome alignment and mitotic spindle organization during their proliferation in the activating LN microenvironment. Disclosure: No conflict of interest disclosed. V665

Novel NO-ASA derivatives are new promising agents in the therapy of chronic lymphocytic leukemia (CLL) and solid cancers Flamme H.1, Krüger M.2, Berkessel A.2, Hallek M.1, Kreuzer K.A.1 Klinik I für Innere Medizin | Universitätsklinikum, Köln, Germany, 2Universität Köln, Department für Chemie, Köln, Germany 1

Introduction: CLL is characterized by an accumulation of B cells in the peripheral blood, bone marrow and lymphatic tissues. Patients harbouring inferior prognostic markers as the 17p deletion or the TP53 mutation show high resistance to standard therapy in CLL. It is well known that NO-ASA (Nitric-oxide donating acetylsalicylic acid) is a potential drug for the treatment of diverse cancers by inducing apoptosis in vitro & in vivo. According to this we developed new NO-ASA derivatives (B9, B12 and B13) which showed lower toxicity in CLL cells compared to healthy PBMCs. Methods: Three outstanding NO-ASA derivatives were tested for their efficacy to induce apoptosis on primary CLL cells with a TP53 mutation and primary CLL cells of high risk patients. The anti-tumor effect analysis of the most promising derivatives (B9, B12, B13) was enlarged to the analysis of diverse cancer cell lines (SW480, MelJuso, HCC44, SH2, COLO407, JIMT1). In addition, the in vivo effect of B1 (para-NO-ASA), B9, B12 and B13 was analyzed via JVM3-xenograft mouse model. To elucidate the biological mechanism of the derivatives we performed a kinase array, Western Blots and immunofluorescence analyses with a fluorescence coupled B9 (B20). Results: We detected a strong reduction of the cell viability in all tested cell lines and a cytotoxic effect even on TP53 mutated and pretreated CLL. The highest effect was seen with B9 and B12. Derivative B9 and B12 demonstrated a strong anti-tumor efficacy in the xenograft mouse with a maximal tumor inhibition rate of 65–70%. The human phosphor kinase array and Western Blot analyses showed preliminary results of the impact of the NO-ASA derivatives on the cell cycle checkpoint signaling and the MAPKinase signaling pathway. After 3h NO-ASA treatment (B9, B12) on primary CLL cells the phosphorylation of the kinases p38 alpha, MKK3/ MKK6, CHK2 and CREB were induced. Conclusion: We showed that our new developed NO-ASA derivatives (especially B9 and B12) are novel potential therapeutic drugs in the treatment of CLL patients including these with bad prognosis. For the first time we showed that the three NO-ASA derivatives B9, B12 and B13 are potential drugs also for diverse solid cancers. Disclosure: Hanna Flamme: No conflict of interest disclosed. Karl Anton Kreuzer: Honoraria: Patent auf die Substanzen.

Abstracts

V666

CLL2-BIG, -BAG, -BCG and -BIO: A series of four phase-II trials evaluating a sequential regimen of combined targeted therapy aiming for a total eradication of MRD in treatmentnaive and relapsed/refractory chronic lymphocytic leukemia (CLL) Cramer P.1, von Tresckow J.1, Bahlo J.1, Engelke A.1, Langerbeins P.1, Fink A.-M.1, Wesselmann J.S.1, Annolleck T.1, Pflug N.1, Kovacs G.1, Fischer K.1, Wendtner C.-M.1,2, Kreuzer K.-A.1, Stilgenbauer S.3, Böttcher S.4, Eichhorst B.1, Hallek M.1, Deutsche CLL Studiengruppe (GCLLSG) Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany, 2Klinikum Schwabing, Klinik für Hämatologie, Onkologie, Immunologie, Palliativmedizin, Infektiologie und Tropenmedizin, München, Germany, 3Universitätsklinikum Ulm, Klinik III für Innere Medizin, Ulm, Germany, 4Universitätsklinikum Schleswig-Holstein, Campus Kiel, Klinik für Innere Medizin II, Kiel, Germany 1

Introduction: The therapeutic landscape in CLL currently undergoes considerable changes with novel antibodies and targeted drugs with exciting efficacy and tolerability becoming available. The GCLLSG proposed the theoretical “sequential triple-T” concept of a tailored and targeted treatment aiming for a total eradication of minimal residual disease (MRD) with a debulking with mild chemotherapy followed by an induction and a MRD-tailored maintenance treatment with an antibody and a kinase Inhibitor or bcl-2-antagonist [Hallek M., ASH 2013]. Methods: Four phase-II trials were designed based on this triple-T concept for 1st-line and relapse treatment of an all-comer population of physically fit and unfit CLL pts. with and without comorbidites. After an optional debulking with 2 cycles bendamustine (which may be omitted in case of a low tumor burden or contraindications), 6 cycles of induction and up to 24 months of maintenance therapy will be administered until achievement of a MRD negative complete remission. Each trial evaluates a different combination of oral drug and antibody: ibrutinib and GA101 (obinutuzumab) in the CLL2-BIG-trial, ABT-199 (venetoclax) and GA101 in CLL2-BAG-, CAL-101 (idelalisib) and GA101 in CLL2-BCG- and ibrutinib and ofatumumab in the CLL2-BIO-trial. Primary endpoint of all trials is overall response rate at the end of induction therapy. However, these trials are not designed for comparisons of the different combinations. 62 pts will be recruited into each of the four multicenter, open-label trials. Results: As of 11th May 2015, 39 pts were included into the CLL2-BIGtrial, 18 for 1st-line and 21 for relapse treatment. In 27 pts a debulking with bendamustine was recommended before induction. Thus far, 10 serious adverse events occurred in 7 pts (4 1st-line and 3 relapsed/refractory): 3 pneumonias, 1 systemic inflammatory response syndrome and 1 postrenal renal failure during debulking as well as 2 infusion-related reactions, 1 thrombocytopenia, 1 pneumonia and 1 episode of upper abdominal pain during induction treatment were reported. The CLL2-BAG and -BCG-trial started recruitment in May 2015 and the CLL2-BIO-trial is expected to start in Q3/2015. Conclusion: These trials investigate different combinations of antibodies and oral targeted drugs as (almost) chemotherapy free, targeted regimen, which are tailored to the pt´s individual disease burden and time point of achievement of total disease eradication (MRD negativity). Disclosure: Paula Cramer: Financing of Scientific Research: honoraria for scientific talks by F. Hoffmann-LaRoche and Janssen-Cilag; Expert Testimony: research funding by F. Hoffmann-LaRoche, Gilead, GlaxoSmithKline and Janssen-Cilag; Other Financial Relationships: travel grants by Astellas, F. Hoffmann LaRoche, Gilead, Janssen-Cilag and Mundipharma Michael Hallek: Advisory Role: AbbVie, Amgen, Celgene, F. Hoffmann-LaRoche, Gilead, Janssen-Cilag and Mundipharma; Financing of Scientific Research: AbbVie, Amgen, Celgene, F. Hoffmann-LaRoche, Gilead, Janssen-Cilag and Mundipharma; Expert Testimony: AbbVie, Amgen, Celgene, F. Hoffmann-LaRoche, Gilead, Janssen-Cilag and Mundipharma; Other Financial Relationships: travel grants by AbbVie, Amgen, Celgene, F. Hoffmann-LaRoche, Gilead, Janssen-Cilag and Mundipharma.

Oncol Res Treat 2015;38(suppl 5):1–270

201


CONTENTS AUTHOR INDEX

Freier Vortrag

Mammakarzinom II / Nierenzellkarzinom V670

No impact of increasing symptom burden on quality of life? – Longitudinal data from the German MaLife-Project on patients receiving monochemo- and endocrine treatment for metastatic breast cancer Marschner N.1, Nusch A.2, Decker T.3, Münz M.4, Kruggel L.4, Jänicke M.4, TMK Registry Group Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg i.Br., Germany, Praxis für Hämatologie und internistische Onkologie, Velbert, Germany, Onkologie Ravensburg, Ravensburg, Germany, 4iOMEDICO, Freiburg i.Br., Germany 1 2 3

Introduction: Treatment of metastatic breast cancer (mBC) aims to prolong survival while maintaining or improving quality of life (QoL). To better understand the real-life situation of patients (pts) in German routine practice, MaLife prospectively investigates patient-reported outcomes during systemic treatment. Patients and methods: MaLife is conducted with the Tumour Registry Breast Cancer II (TMK II), an open, prospective, multicenter, observational study of pts recruited at the start of (neo)adjuvant or palliative systemic therapy in Germany. Besides documentation of medical data, pts regularly receive a set of questionnaires. In total, 2000 pts will be recruited by more than 100 sites. Here, data on global QoL (FACT-G) and symptoms (EORTC QLQ-BR23 – BRST-Score, FACT-Taxane and Brief Fatigue Inventory) are compared during the first 6 months (mts) of mBC treatment with monochemo- (monoCT, n = 166) or endocrine therapy (ET, n = 95). Results: Pts were ø 67/60 (monoCT/ET) years old at the start of therapy. Questionnaire return rate was 80/82% and 64/68% after 3 and 6 mts (monoCT/ET). At the 6 mts survey, 64% of patients continued 1st-line therapy, 23% had changed treatment strategies. Change in treatment and return rate did not affect the overall data presented. About 50% of pts receiving monoCT reported clinically relevant increased symptoms, especially dysgeusia, irritated eyes, hair loss and polyneuropathy after 3 mts of treatment. While polyneuropathic symptoms remained at 6 mts, overall symptom burden decreased. Mean impairment of daily life by fatigue increased in the first 3 mts, but had decreased at 6 mts. About 30% of pts receiving ET reported clinically relevant increased symptom burden at 3 mts, mainly hot flushes, irritated eyes and hair loss. At 6 mts, 30% of pts again reported increasing hot flushes. Mean impairment of daily life by fatigue increased at 3 and 6 mts. Mean global QoL remained unchanged during the first 6 mts of monoCT or ET, and did not differ between the two treatment strategies. Conclusions: Symptoms increase during the first 3 mts of 1st-line treatment and differ between pts receiving monoCT or ET for mBC. In contrast, no change in mean global QoL can be detected. Our data indicate that global QoL alone might not be sufficient to compare treatments and should not serve as the sole base for decision making. Disclosure: No conflict of interest disclosed. V671

Characterization of breast cancer spheroids – ability for generation and marker expression Fröhlich K.1, Haeger J.-D.2, Heger J.1, Pastuschek J.1, Yan Y.1, Photini S.M.1, Lupp A.3, Pfarrer C.2, Markert U.R.1, Schmidt A.1

liferative gradients causing cell heterogeneity in mature spheroids. Three different regions, comprising an outer zone of proliferating cells followed by an inner hypoxic area with quiescent cells and a necrotic core, exist at least. In this study three different breast cancer cell lines were used for optimization of spheroid generation protocols. Further, immunohistochemical markers were tested to characterize the differentiated structure of the spheroids. Methods: MCF-7, MDA-MB-231 and SK-BR-3 cells were cultivated by applying hanging drop, liquid overlay or suspension culture techniques over a time period of two to four days. Experimental approaches differed in cell numbers (400–10,000), media and additives (25% methocel, 25% methocel plus 1% Matrigel, 3.5% Matrigel). In total, 42 different experimental setups were tested. Generation of spheroids was evaluated by light microscopy and the structural composition was assessed by means of Ki67, cleaved poly (ADP-ribose) polymerase (PARP), hypoxia inducible factor 1a, p27 Kip1 and mucin-1 expression. Results: MCF-7 cells formed compact spheroids with almost all methods tested. The fastest and most reliable spheroid generation was obtained by using the hanging drop method with 25% methocel as additive. Here, a clear-cut subdivision into three different regions was detected by Ki-67, cleaved PARP and p27 Kip1. MDA-MB-231 cultivation mostly resulted in the formation of aggregates without forming a spheroidal shape. Only one single protocol (liquid overlay technique, 3.5% Matrigel) led to spheroid formation. None of the protocols yielded SK-BR-3 spheroid formation, while aggregate development was observed under certain conditions. Conclusion: Breast tumor cell lines diverge in their capacity of forming spheroids. Different cell lines require different protocols which cannot simply be transferred from one model to another. In general, we consider hanging drops as an appropriate method for the generation of uniform spheroids with regard to costs, time and experimental handling. Other techniques can be used for cell lines, which have failed to generate spheroids in the hanging drop method. Immunohistochemical analysis should be used as favorable method to prove compact spheroid generation. Disclosure: No conflict of interest disclosed. V672

Quality of life in patients with breast cancer. Results from analysis of our study Qualife and our cancer registry – first analysis Potenberg J.1, Schulz L.2, Linke R.2, Meier F.M.2, Koch A.2, Kirchhoff A.2, Dombrowski-Lütcke M.2, Sprossmann-Günther G.2 Ev. Waldkrankenhaus, Onkologisches Zentrum, Berlin, Germany, Ev. Waldkrankenhaus, Berlin, Germany

1 2

Introduction: Stage and biological factors of breast cancer influence the survival and maybe the quality of life of the patients. Recent studies showed a median overall survival for stage IV patients up the 5 years. The reality in our centre might be different. Therefore we ask the following questions: How are PFS and OS? Is there a difference in the quality of life and in the presence of symptoms according to stage, risk factors and age estimated by our study Qualife? Methods: 2004 in our centre 421 patients were diagnosed with breast cancer, including 48 carcinoma in situ. In the Qualife study 68 patients were evaluated for ECOG, existing symptoms (according CTCAE) and patients were ask to tell her Qol according a numerical scale. Results: Fig. 1 shows the overall survival for the different stages after 5 years.

Friedrich Schiller University, Placenta Laboratories, Department of Obstetrics, Jena, Germany, 2University of Veterinary Medicine, Department of Anatomy, Hannover, Germany, 3Friedrich Schiller University, Institute of Pharmacology and Toxicology, Jena, Germany 1

Introduction: Spheroids display a variety of features, which are absent in cell monolayers. They possess a complex network of cell-cell contacts and advanced extracellular matrix development, as well as metabolic and pro-

202

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V673

Randomized phase 2 three-arm trial of lenvatinib (LEN), everolimus (EVE), and LEN + EVE in patients (pts) with metastatic renal cell carcinoma (mRCC) Motzer R.1, Hutson T.2, Glen H.3, Michaelson M.D.4, Molina A.5, Eisen T.6, Jassem J.7, Zolnierek J.8, Maroto P.9, Mellado B.10, Melichar B.11, Tomasek J.12, Kim H.-J.13, Wood K.14, Dutcus C.13, Larkin J.15 Memorial Sloan-Kettering Cancer Center, New York, United States, 2Texas Oncology, Dallas, United States, 3Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom, 4Massachusetts General Hospital Cancer Center, Boston, United States, 5Weill Cornell Medical College-New York Presbyterian Hospital, New York, United States, 6Addenbrooke’s Hospital, Cambridge Biomedical Research Centre, Cambridge, United Kingdom, 7Medical University of Gdańsk, Gdańsk, Poland, 8Centrum Onkologii, Instytut w Warszawie, Warsaw, Poland, 9Departamento de Oncología Médica, Barcelona, Spain, 10 Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, Spain, 11Onkologicka klinika, Lekarska fakulta Univerzity Palackeho a Fakultni nemocnice, Olomouc, Czech Republic, 12Masaryk Memorial Cancer Institute, Masaryk University, Brno, Czech Republic, 13Eisai Inc., Woodcliff Lake, United States, 14Eisai Limited, Hatfield, United Kingdom, 15The Royal Marsden NHS Hospital, London, United Kingdom 1

Fig. 1.

Fig. 2. Shows correlation between ECOG and the numerical estimation of Qol. Tab. 1 shows that constipation, insomnia, heart problems, dyspnoea and pain were more severe in stage IV patients and insomnia was present in all stages.

symptom

UICC 0 n = 2

UICC I n = 38

UICC II n = 20

UICC III n = 6

UICC IV n = 2

constipation

0

0

0

0

1.5

insomnia

1

1

1.15

1.3

3

paresis

0

0

0

0.8

0

paraesthesia

1

0

0

0.5

0

heart symptoms

0

0

0

0

2

dyspnoea

0

0

0.5

0

2

vertigo

0

0

0.7

0

0

Conclusions: PFS and OS are quite different in a breast centre caring also for older patients when compared with results from recent studies. The numerical scale of Qol correlates with the stage of breast cancer. Constipation, insomnia, heart symptoms, dyspnoea and pain are more common in stage IV patients. Disclosure: No conflict of interest disclosed.

Background: Lenvatinib (LEN), an oral tyrosine kinase inhibitor of VEGFR1–3, FGFR1–4, PDGFRα, RET, and KIT, in combination with EVE had manageable toxicity and antitumor activity in a phase 1 mRCC trial (CCP 2013;73:181). This phase 2, open-label, multicenter study compared LEN, EVE, and LEN+EVE in pts with mRCC. Methods: Pts with progressive clear cell mRCC following 1 VEGF-targeted therapy were randomized 1:1:1 to LEN (24 mg/d), EVE (10 mg/d), or LEN+EVE (18+5 mg/d) in 28d cycles. The primary objective was progression-free survival (PFS) of LEN+EVE or LEN vs EVE. Secondary objectives included overall survival (OS), objective response rate (ORR), and safety. Primary analysis data cutoff was June 13, 2014. Results: 153 Pts were enrolled: 99% had 1 prior VEGF-targeted therapy, 1% had 2; 18% had prior immunotherapy. LEN+EVE prolonged PFS vs EVE (Table; hazard ratio [HR] 0.40; 95% confidence interval [CI] 0.24– 0.68; P< 0.001). LEN alone also prolonged PFS vs EVE (HR 0.61; 95% CI 0.38–0.98; P = 0.048). LEN+EVE and LEN improved ORR vs EVE (P< 0.001 and P = 0.007, respectively). Median duration of response (months) was longest in LEN+EVE, 13.1; LEN, 7.5; EVE, 8.5. OS analysis showed a trend favoring LEN+EVE vs EVE (HR 0.55; 95% CI 0.30–1.01; P = 0.062); this reached significance (HR 0.51; 95% CI 0.30–0.88; P = 0.024) in an updated analysis on Dec 10, 2014. For LEN+EVE, most common anygrade treatment-emergent adverse events (TEAEs) were diarrhea (84%), decreased appetite (51%) and fatigue (47%). Most common grade ≥3 TEAEs were diarrhea (20%), hypertension (14%), and fatigue (10%). Conclusions: LEN+EVE improved PFS and ORR vs EVE alone in this phase 2 trial of pts with mRCC following prior VEGF-targeted therapy. Updated OS also showed improvement with LEN+EVE. A phase 3 randomized trial of the combination in mRCC is planned. Tab. 1.

Primary analysis

LEN+EVE n = 51

LEN n = 52

EVE n = 50

Median survival, months (95% CI)

PFS

14.6 (5.9–20.1)

7.4 (5.6–10.2)

5.5 (3.5–7.1)

OS

25.5 (20.8–25.5) 18.4 (13.3-NE)

17.5 (11.8-NE)

ORR, n (%)

22 (43)

14 (27)

3 (6)

Median duration of response, months (95% CI)

13.1 (3.8-NE)

7.5 (3.8-NE)

8.5 (7.5–9.4)

Median # of cycles, (range)

9.0 (1–25)

8.5 (1–25)

5.0 (1–22)

NE, not evaluable

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

203


CONTENTS AUTHOR INDEX

Disclosure: Robert Motzer: Advisory Role: Pfizer Inc; Expert Testimony: Pfizer, GSK, Novartis, Exelixis, GMS, Genentech, Eisai. James Larkin: Expert Testimony: Novartis, Pfizer, BMS, MSD. V674

Survival data from patients with advanced or metastatic renal cell carcinoma in routine practice differs significantly from clinical trial data – Analyses from the German clinical RCC Registry Marschner N.1, Müller L.2, Staehler M.3, Nusch A.4, Münz M.5, Koska M.5, Jänicke M.5, Goebell P.J.6, and the RCC Registry group Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg i.Br., Germany, Onkologische Schwerpunktpraxis Leer – Emden, Leer, Germany, 3Klinikum der Ludwigs-Maximilians-Universität München Campus Großhadern, München, Germany, 4Praxis für Hämatologie und internistische Onkologie, Ratingen, Germany, 5iOMEDICO, Freiburg i.Br., Germany, 6AURONTE – Urologische Klinik und Klinik für Hämatologie und Internistische Onkologie (Med 5), Universitätsklinik, Erlangen, Germany 1 2

Introduction: Data from (clinical) registration trials are based on patients (pts) who met a number of strict inclusion criteria for eligibility. Many “real life” pts, such as pts with comorbidities, do not fulfil these criteria and are therefore excluded from these trials. In contrast to clinical trials data from registries permit analysing the clinical course of these pts. This allows investigation of potential differences in outcomes like overall survival (OS) between controlled clinical trials and the entire patient cohort. Methods: The prospective German registry on renal cell carcinoma (RCC Registry) includes “all” pts with advanced or metastatic renal cell carcinoma at initiation of systemic first-line therapy. Starting in December 2007, more than 277 office-based medical (uro-) oncologists in Germany are reporting data in the registry. Among other information, data on all systemic therapies, outcome as well as patient and tumour characteristics are collected. We investigated the impact of common exclusion criteria on OS using a multivariate cox proportional hazards model and compared OS of pts potentially eligible or ineligible for clinical trials (interim analysis 15.05.2014). Results: At the time of analysis, data of 732 prospectively documented pts were evaluable to investigate criteria that would determine registration trial eligibility. In a multivariate analysis, the following criteria had a significant negative impact on OS: low Karnofsky performance status (< 80%) (HR 1.745, 95% CI 1.366–2.230, p < .001), low haemoglobin (< lower limit of normal) (HR 1.667, 95% CI 1.348–2.062, p < .001) and nonclear cell carcinoma (HR 1.427, 95% CI 1.123–1.812, p < .01). For 56% of pts at least one of these criteria was documented (“trial ineligible”). For 44% of pts none of these exclusion criteria were present (“potentially trial eligible”). Median OS of “potentially trial eligible” pts was 26.0 months (95% CI 22–32 months) whereas median OS of “trial ineligible” pts was 12.8 months (95% CI 10–16 months). Conclusion: Our data show that more than half of the pts in German routine practice would potentially be ineligible for inclusion in a registration trial. Their OS is inferior compared to “potentially trial eligible” pts. Survival times reported in a registration trial cannot be directly transferred to pts excluded from these trials. Thus physicians should be cautious in interpreting prognosis in routine pts. Disclosure: No conflict of interest disclosed.

V675

Development of a Web-based therapy algorithm tool for renal cell carcinoma (TAT-RCC): Interdisciplinary initiative of NID (Nierenzellkarzinom im Dialog) to ensure a rational work-flow for such a complex and multi-modal malignancy Lüdecke G.1, Doehn C.2, Goebell P.J.3, Grünwald V.4, Kirchner H.5, Overkamp F.6, Schleicher J.7, Schrader M.8, Siebels M.9, Siemer S.10 Universitätsklinikum Gießen und Marburg GmbH, Klinik und Poliklinik für Urologie, Kinderurologie und Andrologie, Giessen, Germany, 2Urologikum Lübeck, Gemeinschaftspraxis, Lübeck, Germany, 3Universitätsklinikum Erlangen, Klinik für Urologie, Erlangen, Germany, 4Medizinische Hochschule Hannover, Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Hannover, Germany, 5KRH Klinikum Siloah, Medizinische Klinik III – Hämatologie und Onkologie, Hannover, Germany, 6 Praxis und Tagesklinik für Internistische Onkologie, Gemeinschaftspraxis, Recklinghausen, Germany, 7Klinikum Stuttgart, Katharinenhospital, Klinik für Hämatologie, Onkologie und Palliativmedizin, Stuttgart, Germany, 8 HELIOS Klinikum Berlin-Buch, Klinik für Urologie, Berlin, Germany, 9Urologie Pasing, Gemeinschaftspraxis, München, Germany, 10Universitätsklinikum des Saarlandes, Klinik fü Urologie und Kinderurologie, Homburg/Saar, Germany 1

Introduction: The renal cell carcinoma is a tumor of high complexity, including tumor-surgery, metastatic-surgery, anti-malignant medical therapy, radiation-therapy and palliative coordination. At each level of this malignancy the decision process has to respect the individual patient reality and the medical facts for optimized therapy to ensure highest rate of curing, live-time prolongation, symptom control and side-effect management. Methods: After an intensive literature triage our interdisciplinary team developed for each therapy level an evidence based mind map to visualize the treatment procedures and their key-points in decision starting from diagnostic techniques over operative procedures to medical treatment in palliative intention including treatment sequence and optimized medical support. The mind mapping technique ensured the easiest way to transfer facts collection into aim achieving decision processes under respect of scientific- and medical evidence. Results: In respect to the individual medical situation of any patient, physicians collect all patient facts to ensure a serious status at the timepoint of interest. With such a collection of information physicians enter TAT-RCC to pick-up information for the next reasonable treatment steps integrating the treatment variability at any level of the decision process. In consequence the TAT-RCC reflects the different operation procedures in respect to organ preservation, renal function and imperative indications, tumor mass reduction, functional recurrence in metastatic tumor and life-time prolongation for isolated metastatic manifestations. Furthermore TAT-RCC incorporated all anti-neoplastic RCC medications at any level in respect to indication, effectiveness and sequence and of course all supportive treatment opportunities for bone problems and pain control. Conclusion: The presented Web-based tool TAT-RCC offer the opportunity to check the reasonable treatment procedures at any level from diagnosis to palliative care. Any physician at any education level will find support to organize an evidence-based treatment in relation to the actual medical condition of his patient. In forensic aspects TAT-RCC is not a medical product but a decision supporting tool with medical freedom based on evidence. It will be available during the congress to all medical users as an open access tool via Internet and in future as an APP for devices. Disclosure: Gerson Lüdecke: Advisory Role: Novartis; Financing of Scientific Research: Novartis, Pfizer, GSK. Stefan Siemer: Advisory Role: Novartis.

204

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Expertenseminar

Ovarialkarzinom fortgeschritten V680

Treatment in advanced ovarian cancer Schönlieb C.1 III. Medizinische Universitätsklinik der Paracelsus Medizinischen Privatuniversität Salzburg, Onkologisches Zentrum Salzburg Cancer Research Institute, Salzburg, Austria 1

Ovarian cancer is the second most common gynecologic malignancy and the most common cause of gynecologic cancer death in the United States. In 2012, 65 583 new cases were diagnosed in Europe. Only 25% of cases are diagnosed in stages I and II. In this meeting we will discuss the optimal treatment strategy for advanced ovarian cancer. Primary surgical cytoreduction followed by systemic chemotherapy has long been the preferred initial management for women with stage III or IV ovarian cancer. In bulky advanced stages IIIC and IV, neoadjuvant chemotherapy with subsequent interval debulking surgery was not inferior to primary cytoreductive surgery followed by adjuvant chemotherapy. Thus, patients with large tumor burden, low albumin levels, or bad general condition are increasingly offered a neoadjuvant approach. For women requiring first-line chemotherapy, the standard intravenous regimen utilizes a combination of a platinum compound and a taxane for six cycles. Nearly 80% of patients achieve remission after first line treatment, but the recurrence rates are 25% for early stage and 80% for advanced stage. When added to standard chemotherapy, the antioangiogenetic drug bevacizumab increases progression-free survival of patients in 1st and 2nd line, and overall survival in subsets of patients. Patient selection, dosage, duration of treatment, and aspects of quality of life are mandatory issues to guide its adequate use. The management of recurrent disease is stratified based upon the time interval between the completion of platinum-based treatment and the detection of relapse, known as the platinum-free interval. Treatment options and the rational for their choice will be discussed. This meeting will discuss the role and sequence of cytoreductive surgery and systemic therapy in 1st and 2nd line, the use of HIPEC, systemic treatment options in platinum-sensitive vs. platinum-resistant recurrent disease, the role of targeted agents like olaparib, and others. Finally two cases will be presented and hopefully interesting cases from the auditorium will be discussed. This meeting should set the stage for an exciting reflection of the current knowledge and a discussion in which we develop optimal care strategies for advanced ovarian cancer.

locally advanced or unresectable pancreatic cancer without measurable metastatic disease. Various combinations of chemotherapy, biologic-targeted therapy, and radiotherapy have been evaluated in different settings to improve outcomes. In this context, a neoadjuvant (preoperative) treatment strategy offers potential benefits: (1) ensuring delivery of early, systemic therapy, (2) improving selection of patients for surgical therapy with truly localized disease, (3) potential downstaging of the neoplasm facilitating a negative margin resection in patients with locally advanced disease. No standard regimen or sequence of treatment could be conclusively determined for neoadjuvant therapy up to now. Current clinical trials investigate chemotherapy regimens like FOLFIRINOX or Gemcitabine/ Nab-Paclitaxel or combination of chemotherapy and radiochemotherapy to improve outcome. Adjuvant therapy consisting of Gemcitabine or 5-fluorouracil over 6 months remains standard of care in Europe after resection of the primary tumor. This has been established in two Phase-III trials. Overall survival showed superior outcome with chemotherapy plus surgical resection versus surgical resection alone. Current clinical trials in adjuvant therapy study the role of additional systemic substances to Gemcitabine as well as the role of radiochemotherapy in addition to Gemcitabine. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Kolon-/Rektumkarzinom II V682

Bevacizumab (bev) combined with chemotherapy as first-line treatment in patients (pts) with metastatic colorectal cancer: Final results from a large non-interventional, communitybased study Hildebrandt B.1, Bruch H.-R.2, Zimber J.3, Hahn L.4, Lück A.5, Timmer H.6, Kroening H.7, Tondar S.8, Kutscheidt A.9, Broszeit-Luft S.10 Charité Universitätsmedizin Berlin, Medizinische Klinik m.S. Hämatologie und Onkologie (CVK), Berlin, Germany, 2Schwerpunktpraxis, Bonn, Germany, 3 Internistische Gemeinschaftspraxis – Praxisklinik, Nürnberg, Germany, 4 Praxisklinik Herne Onkologisches Therapiezentrum, Herne, Germany, 5 Zentrum für Onkologie und Urologie in Rostock, Rostock, Germany, 6 Gemeinschaftspraxis für Hämatologie und Onkologie, Münster, Germany, 7 Gemeinschaftspraxis für Hämatologie und Onkologie, Magdeburg, Germany, 8 Roche Pharma AG, Grenzach-Wyhlen, Germany, 9WiSP Wissenschaftlicher Service Pharma GmbH, Langenfeld, Germany, 10Onkologische Schwerpunktpraxis Dr. S. Broszeit-Luft, Lehrte, Germany 1

Pancreatic adenocarcinoma remains the fourth leading cause of cancer mortality in Europe. The annual incidence of pancreatic cancer is rising with approximately 86,000 new cases and nearly 81,000 patient deaths in 2014. Surgical resection is currently the only treatment option that offers the potential of long-term survival. However, only 20% of patients with pancreatic cancer are candidates for resection. Another 30% of patients have

Background: The safety and efficacy of combined bev+CT in pts with metastatic colorectal cancer (mCRC) in first line setting is consistently proven by randomized clinical trials. This observational trial, conducted in 439 German centers, should evaluate safety and efficacy of bev+CT combinations in daily routine. Methods: Adult patients (>18 years, no upper limit) with histologically proven mCRC scheduled for bev-containing first-line chemotherapy were included if they had no prior palliative pretreatment and no contraindications against bev. Choice of the CT schedule was at the physicians’ discretion. Predefined endpoints included PFS, OS (primary endpoints) and furthers (e.g. treatment characteristics, adverse drug reactions (ADR)). The protocol was amended in March 2013 for the description of efficacy parameters according to the KRAS-status. Results: 3029 eligible pts (female: 37%) were enrolled between 2008 and 2012. Median age was 67 years (range 20–99). 564 pts (19%) were ≥75 years of age. ECOG performance status was favorable (0–1) in 89% of pts. 30% had received previous adjuvant CT. KRAS status (exon 2, Codons 12+13) was available for 57% (n = 1724) pts, mutations were present in 46%. Bev was applied with either fluoropyrimidine (FP) alone (14% in the entire population, 37% in the elderly), FP/irinotecan (49%), or FP/ oxaliplatin (34%). Severe ADRs led to treatment abort in 10% of pts. Spe-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed.

Expertenseminar Pankreaskarzinom V681

Pancreatic cancer: Neoadjuvant and adjuvant therapy Eisterer W.1 Medizinische Universität Innsbruck, Univ.-Klinik für Innere Medizin V, Innsbruck, Austria 1

205


CONTENTS AUTHOR INDEX

cific bev-related ADR were reported within the expected range (gastrointestinal perforation (1%, n = 21), arterial thrombotic events (3%, n = 91) and reversible posterior leukoencephalopathy (<1%, n = 1)). Overall best objective response (PR/CR) was 46% in the entire population (34% in the FP only and 48% in the each triple combination cohort). Based on 2461 observed events (81%), median PFS was 10.3 months (10.0–10.7) for all pts. Median OS (1820 deaths, 60%) was 23.2 months (22.2–24.1) in the ITT population and 19.1 months in the elderly group. The KRAS status was not predictive for both PFS and OS (24.8 vs 23.0 months in favour of wild type, p = 0.24), but the prognostic score according to Köhne was discriminative (p < 0.0001) for both. OS medians were 13.2, 22.1 and 25.4 months in the high, intermediate and low risk groups, respectively. Conclusions: The reliable antitumor efficacy and favorable safety profile of bev+CT could be confirmed in this large observational study, also for the subset of elderly pts. KRAS status was not predictive for survival in this setting. Disclosure: Bert Hildebrandt: Financing of Scientific Research: Roche Pharma AG, Merck, Sanofi, Amgen, Bayer; Expert Testimony: Roche. Stefanie Broszeit-Luft: No conflict of interest disclosed. V683

Treatment results in a community-based colorectal cancer center in Germany – a retrospective analysis of the years 2005–2012 Schmidt M.1, Stumpp C.2, Brinckmann F.2, Heidemann E.3, Mayer J.4, Hebart H.1 Stauferklinikum Schwäbisch Gmünd, Innere Medizin, Schwäbisch Gmünd, Germany, 2Onkologischer Schwerpunkt Stuttgart, Stuttgart, Germany, 3Diakonie Klinikum Stuttgart, Innere Medizin, Stuttgart, Germany, 4Stauferklinikum Schwäbisch Gmünd, Chirurgische Abteilung, Schwäbisch Gmünd, Germany 1

Introduction: The implementation of a nationwide guideline-based therapy in certified cancer centers is necessary to achieve an improvement in patient outcomes. The intention of the study was to critically analyse treatment results in a community-based colorectal cancer center in Germany. Methods: The data of 356 patients with colon cancer (CC) and 191 patients with rectal cancer (RC) have been analysed from Jan 2005 till Dec 2012. Primary endpoint was disease free survival (DFS), disease specific survival (DSS) and overall survival (OS).The treatment results of this study have been compared with international published studies and the published results of the “Onkologischer Schwerpunkt Stuttgart e.V.“ (OSP), that analysed data of 5321 patients with CC and RC from 2004 till 2011. Results: The median age was 74 years for CC and 71 years for RC. Almost a quarter of the patients suffered from metastatic disease (CC 24,9%, RC 23%) at the time of diagnosis. The application of adjuvant chemotherapy (CT) in stage III CC was implemented in 60,2% of the cases. Neoadjuvant or adjuvant chemoradiation was applied in 72,9% of the cases with RC stage II or III. 5-year DSS was 59% for CC and 59,9% for RC. Stage (CC and RC, p < 0,0001), gender (CC, p = 0,0038), age (CC, p = 0,0543), preoperative CEA value (CC and RC, p < 0,0001), appearance of a disease recurrence (CC p = 0,001, RC p = 0,0001, adjuvant CT in stage III CC (p = 0,0127) and resection of metastases (CC p < 0,0001, RC p = 0,0071) influenced the DSS. Compared to the OSP Stuttgart, patients of the Stauferklinikum are more frequently in a locally advanced or metastatic stage and older (CC+3 years, RC+4 years). Adjuvant CT in stage III CC (60,2% vs. 53,1%) and chemoradiation in stage II or III RC (72,9% vs. 59,6%) were applied to a comparable number of patients. 5-year OS was slightly lower (CC 51,3% vs. 56,1%; RC 50% vs. 58,7%), but 5-year DFS was comparable to the results of the OSP Stuttgart (CC 79,6% vs. 79,8%; RC 79,6% vs. 70,8%). Conclusions: The study demonstrates that the guideline-based therapy is used at a high rate in our community-based cancer center at the Stauferklinikum. 5-year DSS, DFS and OS were comparable to international published studies. The lower survival compared to the OSP Stuttgart is

206

Oncol Res Treat 2015;38(suppl 5):1–270

probably due to the higher proportion of high-risk and elderly patients in the Stauferklinikum. Especially for elderly patients, who still have a poor prognosis, it is imperative that new treatment approaches are developed. Disclosure: No conflict of interest disclosed. V684

Panitumumab as first and second line combination therapy of patients with RAS wild-type metastatic colorectal cancer (mCRC): Interim results of the first community-based, observational study Hebart H.1, Tessen H.2, Linde H.3, Koukakis R.4, Rieth A.5, Hellebrand E.5, Reichert D.6 Stauferklinikum Schwäb. Gmünd, Center of Internal Medicine, Mutlangen, Germany, 2Outpatient Clinic, Goslar, Germany, 3Outpatient Clinic, Potsdam, Germany, 4Amgem LTD., Uxbridge, United Kingdom, 5Amgen (Europe) GmbH, München, Germany, 6Outpatient Clinic, Westerstede, Germany 1

Background: Panitumumab (pmab) is approved in Europe for combination with 1st and 2nd line chemotherapy of mCRC patients with RAS wild-type (WT) status. This study evaluated the use of pmab plus chemotherapy in routine clinical practice in Germany. Methods: Participating investigators were asked to prospectively document outcomes and use of pmab as adjunct to either FOLFOX as 1st line therapy (FLT) or to FOLFIRI as second line therapy (SLT) of adult mCRC patients with KRAS (until August 2013) or RAS WT status. Data were collected at 3 time points: baseline (could be done retrospectively for up to 84 days), after treatment had started, and once it was stopped or the 12-month observation period had ended. Results: From Dec 2012 until 15 Jan 2015, 102 patients had received combination treatment with pmab in this ongoing study, 80 as FLT with FOLFOX and 22 as SLT with FOLFIRI. The majority of patients were men ≥65 years with synchronous mCRC and metastases primarily in the liver; 89 subjects had RAS WT and 13 KRAS WT status (without testing for RAS WT). Overall, 81% of patients had undergone prior surgery, most of them with palliative intent. For half of the patients no treatment goal was specified, for the remaining ones tumour shrinkage was most common (25% of FLT patients). On average, patients had received 9 pmab doses (range 1–27) for up to 17 months. Differences in exposure between patient groups were minor, but FLT patients required dose reductions or delays more frequently, particularly due to adverse events (AEs): 25% vs. 9.1%. At the end of the observation period, disease had progressed in 15 FLT (19%) and 4 SLT (18%) patients, of whom 6 and 2 had died, respectively. Overall, 57 FLT (71%) and 12 SLT patients (55%) had AEs, which led to discontinuation in 6 FLT (7.5%) and 3 SLT (14%) patients. In 15 FLT (19%) and 2 SLT (9.1%) patients, AEs were of grade ≥3 and 4 FLT patients had serious AEs. More data will be available at the congress. Conclusions: Pmab is used more frequently in first than in second line combination therapy of mCRC. Good tolerability in general and rate of non-response so far appear to be consistent with data from controlled clinical trials. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V685

V686

Prospective, multi-center study of pharmacokineticallyguided dosing of fluorouracil (5-FU) in metastatic colorectal cancer patients treated with weekly or biweekly 5-FU/ oxaliplatin containing regimens

Exploring the HLA-ligandome of colorectal carcinoma for the development of a personalized immunotherapy

Jaehde U.1, Kunzmann V.2, Link K.3, Holdenrieder S.4, Bertsch T.3, Müller L.5, Ko Y.-D.6, Stötzer O.J.7, Suttmann I.8, Braess J.9, Roessler M.10, Moritz B.10, Kraff S.1, Fritsch A.1, Miller M.C.11, Salamone S.J.11, Wilhelm M.3, CESAR Central European Society for Anticancer Drug Research – EWIV Institute of Pharmacy, Clinical Pharmacy, Universität, Bonn, Germany, Universitätsklinikum, Würzburg, Germany, 3Paracelsus Medical University, Nürnberg, Germany, 4Universitätsklinikum Bonn, Institute of Clinical Chemistry and Clinical Pharmacology, Bonn, Germany, 5Onkologische Schwerpunktpraxis, Leer, Germany, 6Johanniter Krankenhaus, Bonn, Germany, 7HämatologischOnkologische Gemeinschaftspraxis, München, Germany, 8Klinikum Dritter Orden, München, Germany, 9Krankenhaus Barmherziger Brüder, Regensburg, Germany, 10CESAR Central Office, Wien, Austria, 11Saladax Biomedical Inc., Bethlehem, United States 1 2

Introduction: Body surface area (BSA)-based dosing of fluorouracil ­(5-FU) leads to significant underexposure (assessed by area under the concentration-time curve, AUC) in the majority of patients. Personalized, pharmacokinetically (PK)-guided dosing of 5-FU can optimize an individual’s exposure, resulting in a higher overall dose intensity with reduced toxicity and improved outcomes. This study was initiated to validate the use of therapeutic drug monitoring (TDM) to personalize 5-FU dosing in metastatic colorectal cancer (mCRC) patients treated in routine clinical practice. Methods: 75 mCRC patients from 8 different medical centers located throughout Germany received up to 6 cycles of infusional 5-FU according to either the AIO (n = 16), FOLFOX6 (n = 26) or FUFOX (n = 33) regimen. Initial 5-FU dosing for all patients was based on BSA. Individual 5-FU exposure (AUC) was determined from a single blood sample drawn during each infusion using a proprietary immunoassay. To achieve a target AUC of 20 to 30 mg•h/L, subsequent doses of infusional 5-FU were adjusted and optimized according to the previous cycle 5-FU AUC. Primary objective was to confirm that 5-FU TDM resulted in an increased proportion of patients achieving target AUC range at cycle 4 versus cycle 1. Secondary objective was to determine whether 5-FU TDM reduced treatment-related toxicity compared to historical observations. Results: Average 5-FU AUC at cycle 1 was 20 + 15 mg•h/L (coefficient of variation,%CV = 75%), with the majority of patients (62%) falling below, and only 32% falling within, the target exposure range. By cycle 4, average 5-FU AUC was 25 + 7 mg•h/L (p = 0.007), with significantly more patients having optimal exposure (55% within target AUC range, p = 0.005). Overall, 53% of patients had their 5-FU dose increased, 21% were decreased, and 25% remained unchanged. Additionally, less 5-FU-related grade 3–4 toxicity (5% diarrhea, 3% nausea, 0% fatigue and 0% mucositis) was observed compared to historical data (12%, 9%, 12%, 15%, respectively). Conclusion: TDM of 5-FU in routine clinical practice resulted in significantly higher 5-FU exposure, more patients achieving optimal dosing, and less 5-FU-related toxicity. The increase in exposure with simultaneous reduction in toxicity demonstrates the potential for TDM in oncology. Disclosure: No conflict of interest disclosed.

Löffler M.W.1,2, Kowalewski D.1, Dengler F.1,3, Backert L.4, Wagner S.2, Kohlbacher O.4, Stevanovic S.1, Königsrainer A.2, Kanz L.3, Rammensee H.-G.1, Haen S.P.1,3 Interfakultäres Institut für Zellbiologie, Abteilung Immunologie, Tübingen, Germany, 2Universitätsklinik für Allgemeine, Viszeral- und Transplantationschirurgie, Tübingen, Germany, 3Medizinische Universitätsklinik Tübingen, Abteilung II für Onkologie, Hämatologie, Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 4Eberhard Karls University Tübingen, Applied Bioinformatics, Center for Bioinformatics, Quantitative Biology Center, and Dept. of Computer Science, Tübingen, Germany 1

Introduction: Despite recent advances in screening and early detection, colorectal cancer (CRC) remains among the main causes for cancer-related mortality. In recent years, cancer vaccines based on tumor-associated peptides (TUMAP) have progressed into advanced clinical evaluation. So far this novel treatment modality is restricted to HLA-A*02 positive patients and therefore to roughly 50% of the European population only due to the required allotype-specificity of peptides. Since CRC is among the cancers with many genetic mutations leading to cancer-related alterations, a large number of tumor-associated HLA-ligands are expected to be identifiable on CRC cells. Here, we aimed to identify TUMAPs on a more diverse HLA-background to develop a peptide warehouse for the vaccination of CRC patients, allowing a more flexible and personalized approach adapted to the individual antigenic landscape. Methods: The HLA-ligandomes of 30 primary CRC samples and corresponding non-malignant tissues were assessed by HLA-immunoprecipitation and masspectrometry. Results were cross-evaluated on source protein level with our in-house database of benign tissues to exclude expression of HLA-ligands in other organs as best as possible. Immunogenicity testing of the identified ligands is currently ongoing. Results: We could identify about 17,000 peptides on CRC and 39,000 peptides on non-malignant tissue derived from 7,500 and 11,500 unique source proteins, respectively. In order to optimize the applicability of these ligands for a vaccine warehouse, we priorized peptides according to their frequencies of presentation and detectability. Only targets presented on >25% of allotype-matched tumors were included. Mapping the HLA-presented antigenome of primary CRC comparatively in a semi-quantitative manner, 40 highly specific CRC-associated HLA-ligands were identified, which were restricted by one of the eight most frequent HLA-allotypes, covering about 95% of the European population. Conclusions: The identification of CRC-specific HLA-ligands with different HLA-restrictions principally allows the development of peptide warehouse based vaccines, which can be individually composed according to the respective HLA-typing and with the flexibility to adjust it to literally every CRC patient. Hence given immunogenicity of peptides, our data warrant an evaluation of such a vaccine in a Phase I/II clinical trial. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Translationale Forschung V687

Development and clinical implementation of a cross-entity cancer genome sequencing program: NCT MASTER Fröhling S.1, Heining C.1, Gröschel S.1, Hutter B.2, Richter D.1, Geörg C.1, Wolf S.2, Stenzinger A.3, Winkler E.1, Jäger D.1, von Kalle C.1, Brors B.2, Weichert W.3, Glimm H.1 NCT, Heidelberg, Germany, 2DKFZ, Heidelberg, Germany, 3Heidelberg University Hospital, Institute of Pathology, Heidelberg, Germany 1

Introduction: For many cancers, outcomes have improved only marginally in recent years. The potential of genomics to improve the management

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

207


CONTENTS AUTHOR INDEX

of distinct cancer types is exemplified by EGFRmut non-small cell lung cancer (NSCLC), BRAFmut melanoma, and KITmut gastrointestinal stromal tumor, which can be targeted by small-molecule kinase inhibitors, as well as the discovery of genetic determinants of clinical benefit from immune checkpoint blockade in melanoma and NSCLC. While mutations in EGFR, BRAF, KIT, and other cancer genes were discovered through systematic studies of histology-based disease categories, comprehensive genetic analyses of individual cases can also be highly informative regarding disease biology and clinical management. Advances in high-throughput DNA and RNA sequencing provide increasingly more accurate information about all somatic alterations in individual cancer genomes at decreasing costs. However, it is challenging to integrate these analyses into the routine work-up of cancer patients. Methods: We have established NCT MASTER (Molecularly Aided Stratification for Tumor Eradication Research), an institutional review board-approved clinical sequencing program for patients below the age of 50 years with advanced-stage cancer across all histologies. The MASTER workflow involves whole-exome sequencing of tumor and matched normal tissue and RNA sequencing of tumor tissue, followed by technical validation of potentially actionable findings and interdisciplinary evaluation by a dedicated “Precision Oncology Tumor Board”. Results: As of May 2015, 185 patients representing a broad spectrum of entities have been analyzed and discussed in the tumor board. Genomic alterations were categorized according to the level of evidence for an association to drug response, and a potential rationale for experimental therapy was identified in 56% of cases. Based on the signaling pathways affected by these alterations and the resulting points of vulnerability, 6 intervention “baskets” have been defined: (1) tyrosine kinases; (2) PI3KAKT-mTORC1 pathway; (3) RAF-MEK-ERK pathway; (4) developmental pathways; (5) DNA damage response signaling; (6) DNA hypermutation. Conclusions: Our experience demonstrates the feasibility of prospective tumor genome sequencing in a clinical setting. Predefined indicators of efficacy during the current observational cohort study will guide subsequent interventional phase 1b/2 studies within individual, genetically defined baskets. Disclosure: No conflict of interest disclosed. V688

The novel deubiquitinase (DUB) inhibitor b-AP15 induces apoptosis and inhibits proliferation in mantle cell lymphoma Kropp K.N.1, Strunz B.1, Kopp H.-G.1, Grünebach F.1, Kanz L.1, Salih H.R.1,2, Rittig S.M.1, Dörfel D.1,2 Universitätsklinik Tübingen, Innere Medizin II – Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Tübingen, Germany 1

Introduction: The therapeutic efficacy of the proteasome inhibitor bortezomib in mantle cell lymphoma (MCL) is well established which has lead to its incorporation in treatment regimes for this disease. Bortezomib impairs proteasome function by inhibition of the 20S core particle. However, proteasome function can also be targeted by inhibition of 19S deubiquitinase (DUB) activity. Recently, the small molecule b-AP15 was identified as a novel DUB inhibitor and was found to inhibit growth of cancer cells in preclinical analyses (D´Arcy et al., Nat Med 2011). Here we studied whether b-AP15 could also serve as novel treatment option in MCL. Methods: The MCL cell lines REC-1, GRANTA-519, JEKO-1, MINO and Z-138 were exposed to escalating concentrations of b-AP15 for different times. Effects on metabolic activity of the malignant cells were assessed using WST assays, and induction of apoptosis was determined by flow cytometry using dual labeling with Annexin-V and 7-AAD. Furthermore, effects on components of the apoptosis machinery were analyzed by western blot. Results: Treatment of MCL cells with b-AP15 resulted in inhibition of metabolic activity as well as induction of apoptosis in a time and dose

208

Oncol Res Treat 2015;38(suppl 5):1–270

dependent manner. This could be attributed to caspase-3 activation in MCL cells and was mirrored by concomitantly enhanced cleavage of the caspase-cleaved poly-ADP ribose polymerase (PARP). Conclusion: Our results indicate that the DUB inhibitor b-AP15 displays substantial anti-tumor activity in human MCL cell lines and suggest that b-AP15 might be a novel therapeutic option in the treatment of MCL that warrants clinical investigation. Disclosure: No conflict of interest disclosed. V689

Cellular microparticles are released from tumor cells under inflammatory stress and associate with plasma coagulation factor X to form PAR-activating complexes Quecke T.1, Plattfaut C.1, Gamperl H.1, Theophil F.1, Schwenke M.1, Gieseler F.1 UKSH Campus Lübeck, Experimentelle Onkologie, Lübeck, Germany

1

Introduction: Recently, we have described that tissue-factor presenting microparticles (MP) can be isolated from human body fluids using annexinV-coated magnetic beads (Gieseler et al.: Cell biology international, 2014). These MPs are not only coagulation-active but are also able to activate the PAR-signaling pathway in tumor cells, which frequently express PAR1 and PAR2. Methods: Here we show that human tumor cells release considerable amounts of MPs under inflammatory stress induced by TGFbeta or TNFalpha. We examined five different ATCC human tumor cell lines (NIH/ OVCAR-3, ovary adenocarcinoma; Panc-1, pancreas duct adenocarcinoma; A549, lung adenocarcinoma; CaCo2, colorectal carcinoma; MDAMB 231, breast adenocarcinoma) and captured the released MPs. We quantified MPs using cytoflow after specific calibration as well as ELISA based MP activity assay. We characterized MPs using TF- and FX-activity assays as well as ERK activation assays using ELISA technology. Results: Most of these tumor-cell derived MPs express tissue factor (TF), as shown in MP-TF activity ELISA, and bind to annexinV. Due to the missing association of plasma coagulation factors in the cell culture, these tumor-cell MPs do not exhibit FX-activity and their potency to activate PAR and induce ERK phosphorylation is low. After completion of MPs with R1 (calcium, FVa and FXa) , they considerably gained PAR-activating potency to the same rage as MPs isolated from patients (shown by ERK phosphorylation with and without R1 and the specific PAR2 inhibitor GB88). Conclusion: Therapeutic implications of these findings can be demonstrated by the inhibition of MP-induced ERK-phosphorylation with rivaroxaban and tissue factor pathway inhibitor (TFPI). Disclosure: No conflict of interest disclosed. V690

Possible therapeutic implications of structural and functional differences of microparticles from ascites of patients with ovarian cancer Plattfaut C.1, Quecke T.1, Gamperl H.1, Schwenke M.1, Rody A.2, Gieseler F.1 UKSH Campus Lübeck, Experimentelle Onkologie, Lübeck, Germany, 2UKSH, Campus Lübeck, Klinik für Gynäkologie, Lübeck, Germany 1

Introduction: The development of ascites is a serious obstacle for patients with ovarian carcinoma. Due to the increased cellular resistance of tumor cells within this inflammatory environment, treatment is difficult, and the finding of more than 500 ml ascites is an independent negative prognostic marker. Methods: We analyzed patients´ ascites samples with focus on cellular microparticles (MP). Due to the presentation of phosphatidyl serines (PS), p-selectines and tissue factor (TF), MPs can interact with tumor cells and contribute to their treatment resistance. Presented TF associates with plasma coagulation factors to form protease-activated-receptor (PAR)

Abstracts


CONTENTS AUTHOR INDEX

activating complexes. The triggering of PAR signaling pathway through small G-proteins and ERK phosphorylation stimulates EMT and migration of tumor cells. MPs were isolated with annexin-V-coated magnetic beads and analyzed with ELISA techniques for their amount and activity of PS, TF, and coagulation factor X (FX). OVCAR3 cells were used for cell based ERK-ELISA. Results: The amount of PS, which correlates with the amount of captured MPs, was 2,38 ± 0,16 nm, MP-associated TF 14,28 ± 13,13 pg/ml. The quantity of MP-associated FX showed a variation of 14,97%, the amount of proteolytic active FXa was 2375,86 ± 615 ng/ml. MP-associated TF and/ or FXa can activate PAR2, resulting in ERK phosphorylation; ascites derived MPs induced high levels of ERK-phosphorylation in OVCAR3 cells, which can be inhibited by specific PAR2 antiagonizing peptide GB88. Proteolytic activity of MP-associated FXa can be inhibited with the direct FXa-inhibitor Rivaroxaban (100nM). Conclusion: The divergence of MP populations from different patients in the amount of associated FXa is interesting and functionally relevant. TF/ FVIIa as well as FXa are a potent activators of PAR2 and therefore able to induce EMT and tumor cell migration. LMWHs through their potency to induce TFPI release as well as Rivaroxaban through its direct FXa-inhibition, are promising candidates for targetted treatments. Our ongoing study investigates possible consequences of structural and functional differences of ascites derived MPs for clinical outcome. Disclosure: No conflict of interest disclosed. V691

Treatment of oral mucositis following high dose chemotherapy and oral GvHD after allogeneic stem cell transplantation using an Ectoin containing hydroxy-ethylcellulose-gel Haibach M.1, Bilstein A.2, Jitschin R.1, Mougiakakos D.1, Krause S.1, Rösler W.1, Mackensen A.1, Gerbitz A.1 Universitätsklinikum Erlangen, Medizinische Klinik 5, Erlangen, Germany, bitop AG, Witten, Germany

1 2

Introduction: Oral mucositis is a common side effect of chemotherapy. Patients frequently suffer from oral inflammation and severe pain requiring hospitalization for parenteral nutrition and pain management. Furthermore, oral mucositis represents a risk factor for the development of infectious complications. Ectoin, a hydrophilic organic compound, is generated from extremophilic bacteria that show an extraordinary resistance against detrimental conditions. The substance is a medicinal product that has been shown to stabilize natural polymers such as proteins or lipid bilayer membranes. Recent clinical trials have shown positive effects of Ectoin in the treatment of conditions with epithelial barrier damage, like atopic dermatitis, allergies or pharyngitis. Based on these data and on our own experience on the use of Ectoin-containing lotion for the treatment of acute graft versus host disease (GvHD) of the skin we used the substance as an experimental treatment for oral mucositis after high dose chemotherapy. Methods: For sufficient adhesion on the oral mucosa we utilized hydroxy-ethyl-cellulose (HEC) gel supplemented with 2% Ectoin powder. This formulation was administered 4 to 6 times daily in case of mucositis grade III° or IV° (n = 6) after high-dose chemotherapy or for the treatment of oral GvHD following allogeneic stem cell transplantation. Photo documentation was performed prior to treatment and approximately 6 days after first administration. One patient who had previously developed oral mucositis grade IV° after high dose chemotherapy was treated prophylactically. Results: All patients reported a relief of symptoms, especially a reduction of pain after 1 to 4 days. Photo documentation revealed an impressive reduction of inflammation in all patients. The patient undergoing prophylactic treatment developed only grade I° mucositis in contrast to previous courses of high dose chemotherapy. Conclusion: In addition to the hydrating effects of HEC-Ectoin-gel, a rapid relief of symptoms and a visible regression of inflammation occurred

Abstracts

within a few days of treatment during ongoing neutropenia. Preventive use of HEC-Ectoin-gel in one patient resulted in downstaging of mucositis from previously grade IV° to grade I° after high dose chemotherapy. Taken together, these preliminary results demonstrate the potential of Ectoin containing oral mouth wash as a new strategy for the treatment and prophylaxis of chemotherapy induced oral mucositis or oral GvHD. Disclosure: Martina Haibach: Expert Testimony: bitop AG. Armin Gerbitz: No conflict of interest disclosed. V692

Anakoinosis: Communicative reprogramming of tumor systems – for rescuing chemorefractory neoplasia Hart C.1, Vogelhuber M.2, Wolff D.1, Klobuch S.1, Ghibelli L.3, Föll J.4, Corbacioglu S.4, Rehe K.4, Haegeman G.2, Thomas S.1, Herr W.1, Reichle A.1 Department of Internal Medicine III, University Hospital, Regensburg, Germany, University, Gent, Belgium, 3Department of Biology, University Roma, Roma, Italy, 4Department of Pediatrics, University Hospital, Regensburg, Germany 1 2

Disruptive technologies, such as communicative reprogramming (anakoinosis) with cellular therapies in situ for treating refractory metastatic cancer allow patient care to accelerate along a totally new trajectory and highlight what may well become the next sea change in the care of patients with many types of advanced neoplasia. Methods: Cellular therapy in situ consists of repurposed drugs, dual transcriptional modulation (pioglitazone plus all-trans retinoic acid or dexamethasone or interferon-alpha) combined with metronomic low-dose chemotherapy or low-dose 5-azacytidine (epigenetic modulation), plus/ minus classic targeted therapy (anti-inflammatory, immune modulatory, angiostatic). The novel therapeutic tools for specifically designing communicative reprogramming within tumor diseases focus on redirecting (1) rationalizations of cancer hallmarks (the diverse physical constitutions of single cancer hallmarks), (2) modular events (validity of systems participators), (3) the ‘metabolism’ of evolutionary processes (the sum of therapeutically and intrinsically inducible evolutionary processes within the tumor compartment) and (4) the holistic communicative context of structures, functions, decision maxims and hubs in the tumor compartment, which determines validity and denotation of tumor promoting communication lines. Results: Published data on cellular therapies in situ (146 patients, age 2 to 83 years) – inducing cellular reprogramming on-site (in situ) – in castration-resistant prostate cancer (Cancer Microenvironment 2014, Lancet Oncol. 2006; World J Urol 2010), pretreated renal clear cell carcinoma (Biomarker Insights 2007), acute myelocytic leukemia, refractory to standard induction therapy (Haematologica 2014), multiple myeloma > 2nd-line (Blood 2012, abstract), chemorefractory multivisceral Langerhans cell histiocytosis or Hodgkin Lymphoma (Br. J. Haematol 2015), outline the possibility for treating refractory metastatic cancer with the hope that this type of reprogrammed communication will be scalable with minimal toxicity and will finally facilitate to exceed the height of the therapeutic bar. Conclusion: Cellular therapy in situ successfully addresses molecular-genetic heterogeneity at metastatic sites, but also extrinsic and intrinsic drug resistance, by concertedly redirecting still preserved and convergent organized communication tools, while been supported by quite different pattern of chromosomal and molecular-genetic aberrations. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

209


CONTENTS AUTHOR INDEX

Fortbildung

Magenkarzinom V694

Perioperative chemotherapy in localized gastric cancer Wöll E.1 St. Vinzenz Krankenhaus Zams, Innere Medizin, Zams, Austria

1

Introduction: Long term survival in gastric cancer even after complete resection is still poor. In stage I disease, 5-year survival rate is 75% but very few patients are diagnosed in this early stage. In stage II and stage III disease, 5-year survival rate after R0 resection is about 45% and 25% respectively. Therefore even loco-regional gastric cancer should be viewed as disseminated disease. Methods: A review of current literature will be given and the advantages and disadvantages of neoadjuvant (preoperative) or postoperative (adjuvant) therapy will be discussed based on large phase III trials. Results: Perioperative chemotherapy is the standard in caucasian patients in Europe. Different chemotherapy protocols are feasible containing 5FU, platin, epirubicine or docetaxel. The best approach for HER2 positive gastric cancer is still under evaluation. Adjuvant chemotherapy in resected gastric cancer shows a survival benefit predominantly in Asian patients. In the US postoperative chemotherapy combined with radiotherapy is still performed after curative resection. This approach however has several disadvantages. Conclusion: Due to the high relaps rate after complete resection and the postoperative morbidity perioperative chemotherapy with the focus on a neoadjuvant approach is important for optimal multimodality treatment in gastric cancer. Disclosure: No conflict of interest disclosed. V696

New trends in the molecular characterization of gastric cancer and treatment with targeted drugs Lordick F.1 Universitäres Krebszentrum Leipzig (UCCL), Leipzig, Germany

1

The efficacy of cytotoxic therapy in gastric cancer (GC) is limited. This is partly due to the genetic complexity and heterogeneity of GC [Lawrence 2013]. New molecular taxonomies have been published previously and four subtypes have been described: Ebstein-Barr-Virus associated, microsatellite instable, chromosomal instable and genomically stable [TCGA 2014]. Future research will show if this classification contributes to the development of better drugs for GC. To date, trastuzumab, a monoclonal antibody (mAB) directed against HER2, is the only drug with proven efficacy against a molecularly defined (HER2 +) subgroup of GC [Bang 2010]. Lapatinib, a small tyrosine kinase inhibitor (TKI) of HER2 and EGFR, was not effective in 1st- and 2nd-line [Satoh 2014]. Ongoing studies are investigating Pertuzumab and TDM-1 both directed against different domains of HER2. Ramucirumab (RAM) is a human mAB which targets VEGFR-2 and has anti-angiogenic activity. RAM improved survival in 2nd-line GC as a monotherapy and in combination with paclitaxel [Fuchs 2014, Wilke 2014]. There is no biomarker to predict benefit from RAM. mABs against EGFR (Cetuximab, Panitumumab) failed to improve survival in randomized controlled trials [Lordick 2013, Waddell 2013]. Subgroup analyses are ongoing: the SYS-STOMACH consortium which is funded by the German ministry of health (BMBF) seeks to identify biomarkers that determine response and resistance to anti-HER2 and anti-EGFR treatment. While in phase-2 promising results for anti-HGF)/anti-MET directed mABs were published [Iveson 2014], two randomized controlled trials addressing this pathway were stopped prematurely due to lack of efficacy [Cunningham 2015, Shah 2015].

210

Oncol Res Treat 2015;38(suppl 5):1–270

Another interesting target in gastric cancer is Fibroblast Growth Factor Receptor (FGFR) dependent signaling. Nintedanib, a FGFR-2 directed TKI, is being investigated in the EORTC. Preliminary activity has been reported for the anti-PD1 immune checkpoint inhibitor pembrolizumab [Muro 2014]. Correlation with PD-1 expression in GC tissue has been observed which requires confirmation. Targeting cancer stem cell-defining signaling, like the STAT-3/Wnt-pathway, is another approach. BBI-608 inhibits transcription of STAT-3 and other cancer stemness genes [Li 2015] and is being investigated in 2ndline GC. A novel druggable target belonging to the claudin tight-junction protein family is also explored in clinical trials [Türeci 2011]. Disclosure: Florian Lordick: Advisory Role: Biontech, Ganymed, Roche, Lilly, Taiho; Financing of Scientific Research: an Arbeitgeber des Autors; Expert ­Testimony: GSK, Fresenius Biotech; Other Financial Relationships: Reisekosten­ unterstützung für Kongresse: Roche, Bayer, Taiho.

Wissenschaftliches Symposium Transplantation V698

Immunregulation after allo-HCT: Signal transduction and miRs as therapeutic targets Zeiser R.1 Freiburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany 1

Steroid refractory acute and chronic GVHD causes high morbidity and mortality in pateints after allogeneic hematopoietic cell transplantation (alloHCT). Insight into GvHD pathophysiology via the use of mouse models has lead to multiple novel strategies to control this complication including micro RNA, common gamma chain inhibition, protection of the intestinal stem cells by R spondin or IL-22, epigenetic therapy and JAK1/2 inhibition. A particular potent approach could be the modification of micro RNA as they have multiple pro- or anti-inflammatory target genes that can be affected simultaneously. Because microRNA-155 (miR-155) regulates activation of the innate immune system, we aimed to determine its function in dendritic cells (DCs) during GvHD. Experiments using miR-155–/– bone marrow chimeric mice receiving allo-HCT and miR-155– /– DCs showed that miR-155 deficiency in the DC compartment was responsible for protection from GvHD. Activated miR-155–/– DCs displayed lower expression of various purinergic receptors and impaired migration towards ATP. Microarray analysis of LPS/ATP-stimulated miR-155–/– DCs revealed a profound MAPK pathway dysregulation and reduced inflammasome-associated gene expression. Consistent with the expression data, we observed reduced ERK activation, caspase-1 cleavage, and IL-1β production in miR-155–/– DCs. The connection between miR-155 and inflammasome activation was supported by the observation that Nlrp3/miR-155 double knockout allo-HCT recipient mice experienced no increased protection from GvHD compared to Nlrp3–/– recipients. This study indicates that during GvHD, miR-155 promotes DC migration towards sites of ATP release accompanied by inflammasome activation. Inhibiting pro-inflammatory miR-155 by antagomir treatment could reduce GVHD in patients. IN a second set of experiments we studied the common γ chain (CD132) which is a subunit of the interleukin (IL) receptors for IL-2, IL-4, IL-7, IL9, IL-15, and IL-21. T cells exposed to anti-CD132 mAb had a more naive phenotype in microarray-based analyses and showed reduced Janus kinase 3 (JAK3) phosphorylation upon activation. Consistent with a role of JAK3 in GVHD, we found that Jak3(–/–) T cells caused less severe GVHD. These two examples indicate that micro RNAs and signaling pathways can be potent targets to interfere with GVHD. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

Freier Vortrag

V706

Hodgkin-Lymphome / B- und T-Zell-Lymphome

Wnt5a signaling mediates cell migration and invasion of Hodgkin Lymphoma in vitro and in xenograft models

V704

Linke F.1, Zaunig S.1, von Bonin F.1, Wilting J.2, Bryja V.3, Pukrop T.1,4, Trümper L.1, Kube D.1

Moraxella catarrhalis – the infectious origin of nodular lymphocyte predominant Hodgkin lymphoma of IgD type? Thurner L.1, Hartmann S.2, Kemele M.1, Regitz E.1, Fadle N.1, Preuss K.-D.1, Bohle R.-M.1, Vornanen M.3, Kempf V.A.J.4, von Müller L.5, Küppers R.6, Hansmann M.-L.2, Pfreundschuh M.1 Saarland University Medical School, José Carreras Center for Immuno- and Gene Therapy and Internal Medicine, Homburg/Saar, Germany, 2Goethe University Hospital of Frankfurt Main, Dr. Senckenberg Institute of Pathology, Frankfurt am Main, Germany, 3Department of Pathology, Tampere University Hospital and University of Tampere, Tampere, Finland, 4Institute of Medical Microbiology and Infection Control, Hospital of the Johann Wolfgang von Goethe University, Frankfurt am Main, Germany, 5Institute of Medical Microbiology and Hygiene, University of Saarland, Homburg/Saar, Germany, 6 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Essen, Germany 1

Background: Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is a rare type of Hodgkin lymphoma. In contrast to Hodgkin-Reed-Sternberg cells in classical Hodgkin lymphoma, lymphocyte predominant (LP) cells, the tumor cells in NLPHL, show functional variable region genes of immunoglobulins with ongoing somatic hypermutation. Chronic B cell receptor (BCR) stimulation has been proposed to play a central role in the pathogenesis of NLPHL. Aim of the study was to identify potential target antigens of B cell receptors of LP cells. Methods: Recombinant Fabs were constructed of corresponding variable region heavy and light chain genes from isolated LP cells and screened for self- and bacterial antigens. Results: Fabs of 13 cases were constructed. Protein array screening for autoantigens resulted in ribosomal protein S27a and pyruvate carboxylase as target antigens of two individual Fabs. When tested on the lysates from 13 bacterial strains, the recombinant BCR of 4 additional patients reacted with the lysate from Moraxella catarrhalis (MC), a common bacterium colonizing the upper respiratory tract, which expresses IgD-binding protein (MID/hag) enabling MC to bind to the Fc part of IgD and thus activate IgD carrying B-cells. Interestingly, the Fabs of the four patients reactive with MC were derived from IgD+ NLPHL, which is known to represent a peculiar clinical subgroup, affecting the cervical lymph nodes of young people with a strong male predominance. Subsequently, DNA-directed RNA polymerase subunit beta´ of MC (rpoC), a protein of 155 kD, was identified as specific antigen of 4/6 rec. Fabs of IgD+ NLPHL. Serum was available from 2 of these patients and contained Anti-rpoC-Antibodies in relevant titers. Conclusions: The association of MC with IgD+ NLPHL strongly suggests a causal role of MC in the pathogenesis of IgD+ NLPHL. MC is capable of binding to the Fc fragment of LP-BCR of the IgD type via its IgD-binding domain and can mediate chronic antigenic stimulation of B-cells with specificity of MC-rpoC. The presence of serum antibodies against MCrpoC in relevant titres in patients with IgD+ NLPHL suggests that NLPHL is the result of a clonal evolution from a polyclonal B-cell response against MC-rpoC. Our findings are fundamental for the understanding of the pathogenesis of this peculiar clinical subtype of NLPHL and might have relevance for the prophylaxis and treatment of this disease. Disclosure: No conflict of interest disclosed.

Abstracts

University Medical Centre of the Georg-August University of Göttingen, Hematology & Medical Oncology, Göttingen, Germany, 2University Medical Centre of the Georg-August University of Göttingen, Anatomy and Cell Biology, Göttingen, Germany, 3Masaryk University, Experimental Biology, Faculty of Science, Brno, Czech Republic, 4University Hospital Regensburg, Internal Medicine III, Regensburg, Germany 1

Introduction: Hodgkin lymphoma (HL) is characterized by a minority of malignant cells embedded in a background of reactive lymphoid stroma. Despite the identification of a complex network of deregulated pathways in HL, their influence on lymphoma migration, invasion, and the impact onto the lymphoma microenvironment has not yet been investigated in detail. Methods: Migration and invasion analysis have been carried out using the modified Boyden chamber assay. For intervening the Wnt pathway chemical inhibitors, siRNA or corresponding overexpression plasmids for different Wnt ligands and Wnt-pathway components have been applied. For the in vivo chorioallantoic membrane (CAM) assay chicken eggs have been used and evaluated using light microscopy, histology and immune-fluorescence staining. Results: The migratory capacity of HL cells is higher compared to aggressive Non-Hodgkin lymphoma cells and specifically depends on Wnt signaling. The inhibition of Wnt-ligand secretion by porcupine inhibitors reduces HL cell migration. This inhibition is rescued with conditioned HL media suggesting an involvement of autocrine Wnt activity. The overexpression of several non-canonical Wnts shows that Wnt5a specifically stimulates HL cell migration. Knockdown of Frizzled5 and the adaptor protein Dvl disrupts the capacity of Wnt5a to stimulate HL cell migration. Wnt5a-mediated activation of Dvl leads to RhoA activation. Furthermore, Wnt5a affects tumor-stroma interactions in the CAM xenograft model thereby affecting tumor outcome and tumor vascularization. Conclusions: Autocrine Wnt signaling is important for HL cell migration and invasion but also affects the interaction of lymphoma cells with their microenvironment in a paracrine manner. Disclosure: No conflict of interest disclosed. V707

Clinical outcome of primary CNS lymphoma comparing the setting of “real-life” with a clinical trial: A single centre experience of 95 patients Zeremski V.1, Fischer T.1, Schalk E.1 Otto-von-Guericke University Magdeburg, Medical Centre, Department of Haematology and Oncology, Magdeburg, Germany 1

Introduction: Chemotherapy with high-dose methotrexate +/– radiotherapy is a standard therapy approach for primary CNS lymphoma (PCNSL). Here we investigated whether results from clinical trials in PCNSL are comparable to results obtained in a “real-life” setting. Therefore, the primary objective was to compare clinical data and outcome of patients treated outside and within a multicentre clinical trial (G-PCNSLSG-1; Lancet Oncol 2010;11:1036) in all consecutively treated patients at our centre within the last 15 years. Methods: We analysed retrospectively all patients with PCNSL treated in our institution between 11/2000 and 04/2015. Characteristics of patients treated outside the trial (R-LIFE group) were compared with patients treated within the G-PCNSL-SG-1 trial (TRIAL group;). For estimation of clinical relevance of differences, effect size Cohen’s d was stated. d = 0.2, d = 0.5, d = 0.8 means a small, medium and large effect, respectively (Prof Psychol Res Pract 2009;40:532). Results: Altogether, 95 patients were analysed (mean 64.4 years, 50.5% male). Completeness of the study was high (Wu’s C*=89.1%). Median

Oncol Res Treat 2015;38(suppl 5):1–270

211


CONTENTS AUTHOR INDEX

follow-up was 5.9 months (range 0.2–170.0 months). Twenty patients were treated within the clinical trial. Majority of patients (n = 82) received high-dose methotrexate as first-line treatment. Overall response rate was 55.7% (n = 39/70) after primary therapy. Median overall survival (OS) and progression-free survival (PFS) of the whole entire population was 9.6 months (95% CI 1.4–17.8) and 3.4 months (95% CI 3.1–3.7), respectively. The R-LIFE patients were older (median age 69 vs. 62 years; p = 0.031, d = 0.50), had a worse ECOG performance status (1.8 vs. 1.0; p = 0.002, d = 0.80) and a worse Karnofsky index (66.1 vs. 79.0; p = 0.03, d = 0.77) as well as a worse MKSCC score (2.3 vs. 2.0; p = 0.047, d = 0.53) than the TRIAL patients. Of note, median PFS was significantly inferior for the R-LIFE patients compared to the TRIAL patients (3.2 [95% CI 2.1–4.3] vs. 24.3 months [95% CI 3.7–44.9]; hazard ratio (HR)=0.52 [95% CI 0.29–0.92]; p = 0.025, d = 1.17). Furthermore, median OS was also significantly shorter for the R-LIFE patients than for the TRIAL patients (7.3 [95% CI 3.1–11.5] vs. 39.0 months [95% CI 15.3–62.7]; HR = 0.48 [95% CI 0.25–0.92]; p = 0.027, d = 1.20). Conclusions: Our data indicate that treatment outcome is still poor for most of PCNSL patients. Improved survival is limited to young and fit patients, which usually show overrepresentation in clinical trials. Disclosure: No conflict of interest disclosed. V708

Combination therapy with crizotinib/Brentuximab Vedotin in chemorefractory ALK-positive ALCL is feasible and highly effective – a case report Stumme H.1, Lang P.2, Woessmann W.3, Ott G.4, Vöhringer M.5, Handgretinger R.2, Hebart H.1 Stauferklinikum Schwaeb. Gmuend, Zentrum Innere Medizin, Schwerpunkt Haematologie/Onkologie, Mutlangen, Germany, 2Universitaetsklinikum Tuebingen, Allgemeine Paediatrie, Tuebingen, Germany, 3Justus-LiebigUniversitaetsklinikum, Paediatrie/Haematologie/Onkologie, Giessen, Germany, 4 Robert-Bosch-Krankenhaus, Pathologie, Stuttgart, Germany, 5Robert-BoschKrankenhaus, Haematologie/Onkologie, Stuttgart, Germany 1

Introduction: ALK-positive anaplastic large cell lymphoma (ALCL) in children and young adults usually are chemosensitive with an eventfree survival of 70–75%. Due to effective relapse therapy overall survival reaches 90%. However, patients with refractory disease / progression during front-line therapy have a poor outcome. New treatment options for refractory ALK-positive ALCL are needed. Case history/diagnostics: After an injury on the right hand a 19 year old male patient presented a swollen lymph node in the right axilla. The patient developed fever, diarrhea, abdominal pain and cholestatic liver disease suggestive of an infection. Further clinical and histological work-up led to the diagnosis of ALK+ ALCL with non-common subtype. Staging revealed generalized lymphadenopathy, bone marrow- and liver infiltration as well as ascites (Stage IV B). The disease has been completely chemorefractory to a prephase of dexamethasone and one course of CHOEP with evidence of persistent fever, progressive hepatosplenomegaly and GI-involvement. Three-weekly Brentuximab Vedotin was started and shortly thereafter combined with crizotinib. The clinical status of the patient gradually improved. Overall, 4 courses of Brentuximab Vedotin and continuous crizotinib were given resulting in a decrease of the tumor burden indicated by decreasing minimal residual disease and measured by quantitative PCR for NPM-ALK in blood without any considerable toxicities. Three months after start of therapy and just prior start of conditioning therapy for a planned allogeneic transplant, a general seizure as sign of meningeosis lymphomatosa occurred. Following intrathecal therapy and a CNS-intensified preparative conditioning regimen, allogeneic stem cell transplantation from a matched unrelated donor has been performed. The patient remains relapse-free until day 150. Discussion: The combination of the ALK inhibitor crizotinib and the anti-CD30 immunotoxine Brentuximab Vedotin proved to be safe and highly effective for remission induction in a patient with chemorefractory ALK-positive ALCL. However, this case illustrates the urgent medical

212

Oncol Res Treat 2015;38(suppl 5):1–270

need for the development of more effective treatment strategies for the CNS compartment. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium

DGTI Joint Symposium: Neuartige Zelltherapien V709

alloCELL: Efficient allocation of antiviral T-cell preparations for timely therapeutic intervention Maecker-Kolhoff B.1,2, Köhl U.2,3, Blasczyk R.2,4, Eiz-Vesper B.2,4 Hannover Medical School, Department of Pediatric Hematology and Oncology, Hannover, Germany, 2Hannover Medical School, Integrated Research and Treatment Center (IFB-Tx), Hannover, Germany, 3Hannover Medical School, Institute for Cellular Therapeutics, Hannover, Germany, 4Hannover Medical School, Institute for Transfusion Medicine, Hannover, Germany 1

Patients after transplantation are rendered susceptible to infections and reactivations caused by cytomegalovirus (CMV), Epstein-Barr virus (EBV), human herpesvirus 6 (HHV6), adenovirus (ADV), and polyoma virus BK (BKV). The shortcomings of conventional therapies have increased the interest in adoptively transferred antiviral T cells. The efficacy of adoptive immunotherapy and the clinical outcome in high risk patients will be improved by a rapid recruitment of a T-cell donor and an established method for fast manufacturing of antiviral T cells. An allogeneic cell registry (alloCELL, www.alloCELL.org) was established at Hannover Medical School currently recording more than 1350 HLAtyped donors being intensively characterized for their antiviral memory T-cell profile. The alloCELL lab further established a comprehensive protocol to consider clinical requirements of patients at high risk for viral infections or with failed conventional antiviral therapy. The manufacturing license was obtained for generating clinical-grade CMV-, ADV-, EBVand multivirus-specific T-cell products according to the German Medicines Act (AMG) using the CliniMACS cytokine capture system (CCS) and overlapping peptide pools. T-cell donors were defined as eligible if ≥0.03% virus-specific IFN-γ+ T cells are detectable by cytokine secretion assay (CSA). A related or at least 3/6 HLA-A/B/DR-matched alloCELL donor is chosen, if the stem cell donor is not eligible. Determination of antiviral T-cell frequencies in patients after transplantation was done routinely by weekly IFN-γ ELISPOT and HLA multimer staining. 20 out of 53 monitored patients from Hannover Medical School and other hospitals failed to respond to antiviral treatment and were selected for adoptive T-cell transfer. For these patients 54 donors were tested: 25 family, 8 stem cell and 21 alloCELL donors. Clinical-grade CMV-, EBV-, ADV- and CMV/ADV-specific T cells were generated from family and alloCELL donors. The T-cell recipients at Hannover Medical School were monitored to determine T-cell frequencies and chimerism. All patients tolerated the antiviral T-cells without significant side effects and remained clinically free from viral disease (>6 months). These data underline the need of (i) accurate monitoring of viral load and antiviral T-cell frequencies in patients, (ii) early selection of suitable T-cell donors and (iii) support data about safe application of third-party antiviral T-cell products. Disclosure: No conflict of interest disclosed. V711

Generating HLA deficient platelets from induced pluripotent stem cells Figueiredo C.1, Blasczyk R.1 Mediziniche Hochschule Hannover, Institut für Transfusionsmedizin, Hannover, Germany 1

HLA-mediated platelet (PLT) transfusion refractoriness remains a significant clinical problem. In these cases, it would be desirable to have PLT units which are devoid of HLA antigens on their surface. Previously, we

Abstracts


CONTENTS AUTHOR INDEX

showed that generating HLA class I-silenced PLTs from CD34+ progenitor cells using a lentiviral vector encoding for a short hairpin (sh)RNA specific for β2-microglobulin transcripts is feasible. We demonstrated that HLA-silenced PLTs are functional in vitro and efficiently protected against anti-HLA antibody-mediated cytotoxicity in vitro and in vivo. However, the limited availability of CD34+ progenitor cells represents a hurdle for large-scale PLT production. We therefore evaluated the feasibility of induced pluripotent stem cells (iPSCs) as an unlimited cell source for HLA-universal PLT production. Differentiation into megakaryocytes (MKs), the direct precursors of PLTs, and PLTs was performed by incubating iPSCs in monolayer culture in the presence of BMP4 and VEGF for 4 days to initiate mesoderm induction followed by incubation with TPO/ SCF/IL-3 for further 20 days. MK and PLTs differentiation from iPSCs was monitored by anti-CD41, CD42a and CD61 antibodies in flow cytometry as well as morphological analysis using fluorescence microscopy. We differentiated HLA-silenced iPSCs into mature MKs and PLTs. Flow cytometric analysis revealed a mean frequency of 85% of cells positive for the megakaryocytic CD41, CD42a and CD61 with capacity to produce HLA-silenced PLT after transfusion into a mouse model. In addition, increase in cell ploidy by up to 32n was observed. Microscopic analysis showed the formation of pro-PLTs. The PLTs harvested from the differentiation cultures expressed CD61, CD42a and CD42b. iPSCs-derived PLTs demonstrated to aggregate well in response to external stimuli. These results demonstrate the efficient differentiation of iPSCs into mature MKs and PLTs. Therefore, iPSCs including genetically modified cell variants are a highly promising cell source for future large-scale PLT production. The option to irradiate the final product completely eliminates all risks associated with iPSC-based therapeutics and genetic engineering making iPSC-derived PLTs an ideal candidate for a safe genetically modified iPSC product for clinical applications. Thus, the provision of HLA-universal PLTs units may become an important component in the management of patients with platelet transfusion refractoriness. Disclosure: No conflict of interest disclosed.

Freier Vortrag

AML experimentell III V714

Transient in vivo DNA demethylation in peripheral blood blasts of AML patients treated with Decitabine Blagitko-Dorfs N.1, Schlosser P.1,2, Claus R.1, Ma T.1, Ziegler R.1, Götze K.3, Heuser M.4, Heil G.5, Weßendorf S.6, Neubauer A.7, Krauter J.8, Dresel I.9, Döhner K.10, Bug G.11, Lindemann H.-W.12, Salih H.R.13, Scholl S.14, Müller-Tidow C.15, Brugger W.16, Crysandt M.17, Egger M.18, Germing U.19, Lange E.20, Giagounidis A.21, Grishina O.22, Hackanson B.1, Schumacher M.2, Lübbert M.1 University Medical Center Freiburg, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 2University Medical Center Freiburg, Center for Medical Biometry and Medical Informatics, Freiburg, Germany, 3University Hospital of Munich, Department of Internal Medicine III, Munich, Germany, 4Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany, 5Klinikum Lüdenscheid, Department of Hematology and Oncology, Lüdenscheid, Germany, 6Klinikum, Esslingen, Germany, 7Philipps University of Marburg, Department of Hematology, Oncology and Immunology, Marburg, Germany, 8Städtisches Klinikum Braunschweig, Department of Hematology and Oncology, Braunschweig, Germany, 9Ortenau-Klinikum Offenburg-Gengenbach, Department of Hematology, Oncology and Palliative Medicine, Offenburg, Germany, 10University Hospital Ulm, Department of Internal Medicine III, Ulm, Germany, 11Goethe University Frankfurt, Department of Hematology, Frankfurt, Germany, 12St.-Marien-Hospital, Klinik für Hämatologie und Onkologie, Hagen, Germany, 13University of Tübingen, Department of Hematology, Oncology and Immunology, Tübingen, Germany, 14Universitätsklinikum Jena, Department of Hematology and Oncology, Jena, Germany, 15Universitätsklinikum Halle, Department of Hematology and Oncology, Halle, Germany, 16Schwarzwald-Baar Klinikum, Med. Klinik II, Onkologie, Hämatologie, Immunologie, Infektiologie und Palliativmedizin, Villingen-Schwenningen, Germany, 17Universitätsklinikum Aachen, Med. Klinik IV, Hämatologie und Onkologie, Aachen, Germany, 18 Ortenau Klinikum Lahr-Ettenheim, Lahr, Germany, 19Universitätsklinikum Düsseldorf, Department of Hematology, Oncology and Clinical Immunology, Düsseldorf, Germany, 20Evangelisches Krankenhaus Hamm GmbH, Medizinische Klinik Hämatologie/Onkologie, Hamm, Germany, 21Marien Hospital Düsseldorf, Klinik für Onkologie, Hämatologie und Palliativmedizin, Düsseldorf, Germany, 22 University Medical Center Freiburg, Clinical Trials Unit, Freiburg, Germany 1

Introduction: Low-dose DNA hypomethylating agents represent a therapeutic option in treatment of elderly acute myeloid leukemia (AML) patients (pts). To unravel the in vivo mechanism of action of these agents for clinical response, we set out to characterize methylomes of leukemic blasts before and during treatment of AML pts enrolled in the randomized phase II DECIDER clinical trial (NCT00867672). Methods: Peripheral blood mononuclear cells from AML pts were collected before and on days 4, 8 and 15 of therapy (i.v. 20mg/m2 decitabine for 5 days, with or without subsequent oral drug add-on ATRA, valproic acid). Leukemic blasts were isolated using magnetic sorting of cells labelled with anti-human CD34 and/or CD117 MACS microbeads. Blast specific genome-wide DNA methylation profiles were obtained using Infinium Human Methylation 450 BeadChip arrays. Data were analyzed using R packages RnBeads applying beta mixture quantile dilation for normalization and a modified version of NHMMfdr for multiple comparisons. Results: Peripheral blood blasts (median purity: 92%) were isolated from 32 pts. In total, until now 71 methylomes were generated and analyzed by longitudinal mathematical modelling. Genome-wide methylation was interpreted blinded to all clinical data including drug add-on. Methylation dynamics were described for each individual CpG and multiple testing was addressed by a highly powerful non-homogeneous hidden Markov model. Across all pts, 15% of CpGs (71,377 of total 462,769 CpGs) became demethylated (Δβ>0.1, FDR< 0.01) by day 8. Strikingly, 82.2% of them (58,706 CpGs) became partially re-methylated by day 15. Most of the demethylated CpGs resided in gene bodies and intergenic regions. The majority of these demethylated CpGs were located in CpG deserts (56.6%), only 8.5% in CpG islands, 20.7% in shores and 14.2% in shelves.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

213


CONTENTS AUTHOR INDEX

Surprisingly, only 6 CpGs became hypermethylated. We are currently performing in silico analysis to determine whether decitabine-”reactive” CpGs are enriched for specific TFBSs, DNaseI hypersensitive sites or particular histone modifications. Conclusions: This first interim analysis revealed significant genome-wide DNA demethylation by day 8, with striking re-methylation by day 15 from start of decitabine treatment in AML blasts in vivo. Remarkably, most of the demethylated CpGs resided in non-promoter regions and in regions with low CpG density. Disclosure: Nadja Blagitko-Dorfs: No conflict of interest disclosed. Michael Lübbert: Advisory Role: Advisory Board for Johnson & Johnson; Financing of Scientific Research: Johnson & Johnson; Other Financial Relationships: received study drug decitabine from Johnson & Johnson, study drug valproic acid from Ratiopharm. V715

The combination of crenolanib and azacitidine effectively targets leukemia-initiating cells in FLT3-ITD+ AML to overcome protection by the bone marrow niche Garz A.-K.1, Weickert M.-T.1, Pagel C.1, Habringer S.1, Vick B.2, Jeremias I.2, Keller U.1, Oostendorp R.1, Peschel C.1, Götze K.1 Technische Universität München, III. Medizinische Klinik, München, Germany, Helmholtz Zentrum, München, Germany

1 2

Introduction: Clinical trials have shown that treatment of FLT3-ITD+ acute myeloid leukemia (AML) with tyrosine kinase inhibitors (TKI) leads to clearance of peripheral leukemic blasts, whereas the bone marrow response is less pronounced and remissions are short-lived. This suggests a protective effect of the marrow niche on leukemic cells. Recently, we demonstrated that long-term FLT3-ITD+ AML stem/progenitors resist apoptosis by TKI through protection by the marrow niche (Parmar et al.: Cancer Res 2011). Hence, FLT3-ITD+ leukemia initiating cells (LIC) persist after TKI treatment and may expand, leading to AML relapse. The hypomethylating agent azacitidine has been shown to effectively prevent or delay full-blown relapse in MRD-positive AML patients after allogeneic stem cell transplantation (Platzbecker et al.: Leukemia 2012). Here, we examined whether stromal resistance of FLT3-ITD+ LIC could be overcome by the next-generation TKI crenolanib alone or in combination with azacitidine treatment. Methods: FLT3-ITD+ human AML cell lines (MV4–11, Molm-13) or primary CD34+FLT3-ITD+ progenitors isolated from AML bone marrow were cultured on the murine embryonic mesenchymal cell line EL08–1D2 which mimics the niche and maintains LIC for a prolonged period of time in vitro. Co-cultures were treated with DMSO, TKI and/or azacitidine for defined periods. Apoptosis, cell cycle and differentiation of AML cells were analyzed by flow cytometry. Clonogenic capacity and frequency of primitive stem/progenitors was probed by standard CFU and LTC assays. Results: Monotherapy with crenolanib was effective in suspension culture but unable to target FLT3-ITD+ LIC cells protected by niche cells in vitro. However, the combination of crenolanib and azacitidine resulted in efficient apoptosis and significantly reduced clonogenic capacity of CD34+FLT3-ITD+ LIC in the presence of stroma. Treatment of stroma with azacitidine before culture with AML cells did not influence protection against crenolanib. Furthermore, soluble stromal factors did not account for TKI resistance. Conclusion: The combination of azacitidine and next generation TKI is a novel promising treatment regimen to overcome niche protection of FLT3-ITD+ LIC cells in AML. Our data suggest that azacitidine induces differentiation of LIC and thereby impairs stromal resistance against TKI. Validation of synergistic effects are currently being tested in vivo by xenotransplantation studies in NSG mice. Disclosure: Anne-Kathrin Garz: No conflict of interest disclosed. Katharina Götze: Financing of Scientific Research: Celgene GmbH.

214

Oncol Res Treat 2015;38(suppl 5):1–270

V716

Molecular dissection of valproic acid effects in acute myeloid leukemia identifies predictive networks Rücker F.G.1, Schlenk R.F.1, Fütterer M.1, Komarica V.1, Döhner H.1, Schmid M.2, Döhner K.1, Bullinger L.1 Universitätsklinikum, Ulm, Germany, 2Medizinische Onkologie und Hämatologie, Zürich, Switzerland 1

Introduction: Histone deacetylase inhibitors (HDACIs) like valproic acid (VPA) display activity in leukemia models and induce tumor-selective cytotoxicity against acute myeloid leukemia (AML) blasts. However, little is known about underlying mechanisms. Methods: To gain additional insight into VPA treatment associated pathway changes, we performed global gene expression and miRNA profiling and integrated in vitro results from myeloid cell line experiments (CMK, HEL, K562, NB4, and HL60) with in vivo data derived from molecularly well-characterized primary AML samples collected within the AMLSG 07–04 trial (NCT00151242) followed by a correlation of our findings with clinical information. Results: Compared to untreated controls, leukemia cell lines treated with VPA (final concentration 1mM) for 48h showed a dosage dependent down-regulation of HDAC2 and up-regulation of UBC8 and acetylated histone H4 protein levels. To further characterize key genes and molecular pathways affected by VPA treatment in vitro, we performed comparative gene expression profiling (comparison of VPA treated vs. untreated cases). This analysis revealed an in vitro VPA response signature enriched for genes/pathways known to be implicated in cell cycle arrest, apoptosis, and DNA repair (e.g. Cell Cycle G1/S Checkpoint, ATM Signaling Pathway, and RB Tumor Suppressor Signaling in response to DNA damage). Following in vivo VPA treatment of primary leukemia patients (serum levels of 50–110mg/l), gene expression changes showed results concordant with the in vitro VPA response signature, which was also significantly enriched despite concomitant chemotherapy (p < .0001). Comparative miRNA profiling revealed VPA-associated miRNA expression changes likely contributing to a VPA-induced reversion of deregulated gene expression with miR-150 and miR-106b being the most significantly down- (2.7-fold) and up-regulated (2.24-fold) miRNAs, respectively. In addition, we were able to define biomarkers, such as CXCR4 and LBH, predicting VPA response in vivo at the time of diagnosis. These could be validated in an independent cohort of AML patients (n = 114) in which CXCR4 and LBH expression was also associated with a higher rate of complete remission and better relapse-free survival. Conclusions: Our data provide new insights into molecular mechanisms of VPA treatment effects in vitro and in vivo and our findings may prove to be useful in predicting AML patients likely to benefit from epigenetic treatment. Disclosure: No conflict of interest disclosed. V717

MN1-Fli1 oncofusion transforms hematopoietic stem cells into acute megakaryoblastic leukemia cells Wenge D.V.1, Felipe Fumero E.1, Angenendt L.1, Schliemann C.1, Schmidt E.1, Berdel W.E.1, Arteaga M.F.1, Mikesch J.-H.1 Uniklinik Münster, Münster, Germany

1

Acute megakaryoblastic leukemia (AMKL) is a rare subtype of acute myeloid leukemia. Prognosis of AMKL depends upon the cause. Detection of t(1;22)(p13;q13) in children with AMKL correlates with poor prognosis whereas AMKL patients with Down syndrome show 10 year survival rates of around 80%. For around one third of all AMKL cases a cause of the disease has not yet been identified. Long-term outcome of those patients especially in adults is poor. The underlying mechanisms of AMKL leukemogenesis are still largely unknown. Recently, an in frame fusion of MN1 and Fli1 genes was detected in a child with AMKL. We sought out to investigate the potential role of this oncofusion in AMKL leukemogenesis. Interestingly, expression of MN1-Fli1 in murine hematopoietic progen-

Abstracts


CONTENTS AUTHOR INDEX

itor cells (HPCs) was sufficient to largely enhance their self-renewal in vitro as assessed via serial replating. Strikingly, MN1-Fli1 mediated transformation generated immature myeloid cells showing AMKL cytomorphology as well as high expression of typical AMKL markers CD41 and CD61. Systematic structure function analyses revealed FLS and 3´ETS domains of Fli1 as decisive domains for AMKL phenotype. Expressing MN1-Fli1 deletion mutants missing one of these two domains in murine HPCs still induced leukemic transformation of the cells but generated leukemic blasts with a cytomorphology strongly resembling leukemic cells derived from MN1 overexpression and with loss of AMKL markers. Our data highlight an important role of MN1-Fli1 in AMKL leukemogenesis and provide a basis for research evaluating the value of this oncofusion as a future diagnostic marker and/or therapeutic target in AMKL patients. Disclosure: No conflict of interest disclosed. V718

In vivo imaging of chemokine receptor CXCR4 expression by PET/MRI in patients with myeloid malignancies Herhaus P.1, Philipp-Abbrederis K.1, Habringer S.1,2, Vag T.3, Höllein A.1, Gerngroß C.3, Müller-Thomas C.1, Schwamborn K.4, Peschel C.1,2, Schwaiger M.2,3, Wester H.-J.2,5, Verbeek M.1, Götze K.1,2, Keller U.1,2 III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität, München, Germany, 2Deutsches Konsortium für Translationale Krebsforschung (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Partnerstandort München, München, Germany, 3Klinik für Nuklearmedizin, Klinikum rechts der Isar, Technische Universität, München, Germany, 4Institut für Pathologie, Klinikum rechts der Isar, Technische Universität, München, Germany, 5Institut für Pharmazeutische Radiochemie, Technische Universität, München, Germany 1

Introduction: C-X-C-motif chemokine receptor 4 (CXCR4) is a G-protein coupled receptor with one single known chemokine ligand, Stromal cell-derived factor-1α (SDF-1α). The CXCR4/SDF-1α- axis is essential for homing and retention of hematopoietic stem cells to the bone marrow (BM), leukocyte trafficking, as well as the activation of pro-survival signaling cascades. In acute myeloid leukemia (AML) its overexpression has been shown to be an independent risk factor for reduced overall survival and progression-free survival. CXCR4-mediated homing of leukemic cells to protective BM niches promotes treatment resistance and plays an important role for the persistence of minimal residual disease. This renders the CXCR4/SDF-1α- axis a promising therapeutic target in AML, and CXCR4-directed agents are currently under clinical investigation. We tested [68Ga]-Pentixafor, a radiolabeled peptide positron emission tomography (PET) tracer selective for human CXCR4, for its suitability for in vivo imaging, and quantified CXCR4 surface expression on blasts in patient-derived samples. Methods: 7 patients with myeloid malignancies (myelodysplastic syndrome (MDS), secondary AML and de novo AML) were imaged with [68Ga]-Pentixafor by PET magnetic resonance imaging (PET/MRI) after signing informed consent in this observational assessment. Maximum standardized uptake values (SUVmax) of the involved BM areas were compared to BM from patients without BM malignancy. Expression of CXCR4 on myeloid blasts of 50 patients (samples derived from BM and whole blood) was determined by flow cytometry (FACS). Results: Expression of CXCR4 in the BM determined by PET was significantly higher in 3 out of 7 patients with myeloid malignancies (all of them presented with AML) as compared to control BM (mean SUVmax 5.81, range 4.5 to 8.2 vs. 2.66, range 1.6 to 4.1) and correlated with abnormal signals determined by MRI. CXCR4 surface expression on myeloid blast as determined by FACS was higher in patients with AML compared to MDS (relative median expression 1.79 ± 1.89 vs. 0.88 ± 0.28). 13/33 patients with AML compared to 3/17 patients with MDS showed CXCR4 + blasts. Conclusion: Imaging of CXCR4 by PET/MRI with [68Ga]-Pentixafor is feasible in patients with myeloid malignancies. Due to the higher expression in leukemic BM compared to control BM and its low uptake in other

Abstracts

organs (e.g. liver, brain, gut), CXCR4 could serve as an attractive endoradiotherapeutic target in selected patients with AML. Disclosure: No conflict of interest disclosed. V719

Effect of TGF-b1 and CXCL12 blocking on apoptosis, proliferation and cytarabine sensibility of acute myeloid leukemia (AML) cells cocultured/cotransplantated with multipotent mesenchymal stroma cells Schelker R.1, Müller G.1, Hart C.1, Klatt S.1, Herr W.1, Grassinger J.1 Regensburg, Innere Medizin III (Hämatologie/Onkologie), Regensburg, Germany 1

Introduction: Multipotent mesenchymal stroma cells (MSC) display a central cellular factor within the bone marrow (BM) niche, contributing to hematopoietic stem cell (HSC) dormancy, maintenance and proliferation via expression of soluble factors like transforming growth factor-b1 (TGF-b1) and the chemokine ligand CXCL12. The hematopoietic stem cell niche is a flexible system that supports physiological as well as aberrant hematopoiesis in a similar manner. In our study, we therefore sought to investigate whether blocking TGF-b1 and CXCL12 signaling influences the proliferation and chemotherapy sensitivity of leukemic cells (LC). Methods: MSC from AML patients (LD) and donors without BM disorders (BD) were obtained from BM aspirates. Subsequently, the phenotypic and functional criteria of MSC were confirmed and co-cultures of AML cells (LC) on MSC were initiated. The effect of CXCL12 and TGF-b1 blockade on proliferation and apoptosis of LC was tested in these co-cultures with and without the addition of cytarabine. Additionally, the engraftment efficiency of MSC, LC and HSC and combinations thereof in NSG mice was analyzed. The influence of CXCL12 and TGF-b1 blocking in combination with cytarabine on engrafted LC is currently being tested. Results: The in vitro blockade of TGF-b1 increased the proliferation of LC while inhibition of CXCL12 showed anti-proliferative effects. Apoptosis was enhanced in both experimental setups, with TGF-b1 suppression being the predominant. Additionally, CXCL12 inhibition resulted in 13% higher cell reduction compared to control vs. 10% after inhibition of TGF-b1. Humanized NSG mice transplanted with MSC showed improved LC engraftment as compared to control mice without MSC (on average 48% vs. 30% human blasts in PB after five weeks). Conclusions: Our data suggest that MSC support growth of LC in vitro and in vivo. TGF-β1 and CXCL12 signaling alters AML cell proliferation and chemotherapy sensibility. These pathways might be an additional therapeutic target for the treatment of AML. Disclosure: No conflict of interest disclosed.

Freier Vortrag MPN I V720

CMML and aCML share mutated genes and can be discriminated from MDS/MPN, U and RARS-T: The mutational landscape of 18 investigated genes in 177 patients Meggendorfer M.1, Haferlach T.1, Haferlach C.1, Kern W.1, Schnittger S.1 MLL Münchner Leukämie Labor GmbH, München, Germany

1

Introduction: Myelodysplastic/myeloproliferative neoplasms (MDS/ MPN) as defined by the World Health Organization (WHO) classification show features of both MDS and MPN. This category includes atypical chronic myeloid leukemia (aCML), chronic myelomonocytic leukemia (CMML), MDS/MPN, unclassifiable (MDS/MPN, U), and refractory anemia with ring sideroblasts associated with marked thrombocytosis (RARS-T). Some share morphological features, but the mutation landscape has not been comprehensively investigated yet. Therefore we ana-

Oncol Res Treat 2015;38(suppl 5):1–270

215


CONTENTS AUTHOR INDEX

lyzed 18 genes for mutations known to occur in myeloid entities in order to discover entity specific mutation patterns. Methods: We investigated 177 patients diagnosed by cytomorphology and genetic studies strictly following WHO criteria: 35 patients were diagnosed as aCML, 58 as CMML, 39 as MDS/MPN, U, and 45 as RARS-T. All patients were analyzed either for whole coding regions or hotspot mutations of 18 genes: ASXL1, BRAF, CALR, CBL, CSF3R, DNMT3A, ETNK1, JAK2, MPL, NPM1, NRAS, KRAS, RUNX1, SETBP1, SF3B1, SRSF2, TET2, and U2AF1. Gene mutations were analyzed by Sanger sequencing, next generation sequencing, melting curve analyses, or gene scan. Results: Mutational patterns in these four entities showed that ASXL1 and TET2 are frequently affected in all entities but significant differences exist: ASXL1 is less frequently mutated in RARS-T (20%) in comparison to aCML (57%; p < 0.001) and CMML (52%; p = 0.001), TET2 is more often mutated in CMML (53%) in comparison to MDS/MPN, U (26%; p = 0.007) and RARS-T (31%; p = 0.031). SRSF2 mutations appear more often in CMML (53%) than in comparison to RARS-T (9%; p < 0.001) and MDS/MPN, U (15%; p < 0.001), SF3B1 mutations prevail in RARS-T (91%) compared to all other entities (aCML: 11%, CMML: 5%, MDS/ MPN, U: 13%; for all p < 0.001). Thus the four entities are affected by mutations in different pathways: i) JAK2/CALR/MPL (JAK/STAT pathway) preponderate in MDS/MPN, U (33%) and RARS-T (53%) as compared to aCML (9%) and CMML (7%; p < 0.001). ii) NRAS/KRAS/CBL (RAS pathway) were more often mutated in aCML (37%) and CMML (52%), as compared to MDS/MPN, U (5%) and RARS-T (9%; p < 0.001). Conclusions: 1. ASXL1 and TET2 are the most frequently mutated genes in MDS/ MPN overlap entities. 2. SRSF2 is most frequently mutated in CMML, but also in aCML. 3. The JAK/STAT pathway is more often affected in MDS/MPN, U and RARS-T. 4. In contrast, the RAS pathway is more often affected in aCML and CMML.

er CRT group (70.0 yrs). Thrombohaemorrhagic and malignancy event rates are displayed in Table 1. 105 patients transformed to myelofibrosis (MF) and 62 to acute leukaemia (AL). In patients who had only ever received either ANA or hydroxycarbamide (HC), the rate of transformation to MF was higher in the ANA vs HC group (0.78 vs 0.17) whereas transformation to AL was higher in the HC vs ANA group (0.22 vs 0). No unexpected side effects were noted. Conclusions: Patients who received ANA were younger than those who received other CRT. The thrombohaemorrhagic event rates were generally low, with differences in venous thrombotic and haemorrhagic event rates between the ANA+other CRT group. Transformation to MF was more frequent in the ANA group while transformation to AL was more common in the other CRT group.

Disclosure: No conflict of interest disclosed.

Disclosure: Martin Griesshammer: Advisory Role: Shire, Novartis, Lilly, Sanofi, AOP; Financing of Scientific Research: Shire, Novartis, Lilly, Sanofi, AOP. Gunar Birgegard: No conflict of interest disclosed.

V721

Treatment of essential thrombocythaemia in Europe: an observational study of 3649 high-risk patients in EXELS Griesshammer M.1, Stegelmann F.2, Mohr A.3, Matzdorff A.4, Besses C.5, Gugliotta L.6, Harrison C.7, Kiladjian J.J.8, Hamdani M.9, Achenbach H.10, Birgegard G.11 Johannes Wesling Klinikum, Minden, Germany, 2University Hospital of Ulm, Department of Internal Medicine III, Ulm, Germany, 3Onkologiepraxis, Hamburg, Germany, 4Caritasklinik St. Theresia, Saarbrücken, Germany, 5Department of Haematology, Hospital del Mar-IMIM, Barcelona, Spain, 6Department of Haematology, ‘L e A Seragnoli’, St Orsola-Malpighi Hospital, Bologna, Italy, 7 Department of Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom, 8APHP, Hôpital Saint-Louis, Centre d’Investigations Cliniques, Paris, France, 9Global Biometrics, Shire Pharmaceuticals, Wayne, United States, 10Research & Development, Shire GmbH, Zug, Switzerland, 11 Department of Haematology, Uppsala University, Uppsala, Sweden 1

Introduction: The observational EXELS study (NCT00567502) describes the largest prospective cohort of high-risk patients with essential thrombocythemia (ET) reported to date. Methods: High-risk patients (≥1 of age >60 years, previous thrombotic event, platelet count >1000×109/L) with ET were enrolled across 13 European countries between 2005 and 2009. Patients were followed for 5 years. The primary objective was safety and pregnancy outcomes of anagrelide (ANA) compared with other cytoreductive therapies (CRTs). Secondary objectives included efficacy, measured by incidence of thrombohaemorrhagic events and platelet reduction. Event rates are presented as number of patients per 100 patientyears exposure and by treatment at time of event. Results: 3649 patients were categorised according to treatment at registration: ANA (n = 804), ANA+other CRT (n = 141), other CRT (n = 2666) and no CRT (n = 38). Median age was lower in the ANA (55.5 yrs) vs oth-

216

Oncol Res Treat 2015;38(suppl 5):1–270

Tab. 1. Event rates

Treatment at time of event

ANA N = 1127

Other CRT N = 2909

ANA+other CRT N = 451

No CRT N = 645

Total thrombohaemorrhagic events

2.75

2.60

2.86

4.91

Arterial thrombotic events

1.63

1.62

2.25

2.74

Venous thrombotic events

0.35

0.57

0.11

1.13

Major haemorrhagic events

0.87

0.49

0.69

1.12

Transformation to:

Myelofibrosis

1.31

0.32

1.27

2.31

Acute leukaemia

0.17

0.33

0.46

2.57

Non-haematological malignancy

0.49

1.35

0.46

1.95

V722

A phase-Ib/II study of ruxolitinib plus pomalidomide in Myelofibrosis: Data from the MPNSG-0212 Trial (NCT01644110) Stegelmann F.1, Bangerter M.2, Grießhammer M.3, Hebart H.4, Heidel F.5, Hochhaus A.6, Möhle R.7, Reiter A.8, Scheid C.9, Kirschbaum R.1, Reim R.1, Sutter U.1, Döhner H.1, Schlenk R.F.1, Döhner K.1 Klinik für Innere Medizin III, Universitätsklinikum Ulm, Ulm, Germany, Hämatologisch-Onkologische Praxis, Augsburg, Germany, 3Klinik für Hämatologie, Onkologie und Palliativmedizin, Klinikum Minden, Minden, Germany, 4Zentrum für Innere Medizin, Stauferklinikum Mutlangen, Mutlangen, Germany, 5Universitätsklinik für Hämatologie und Onkologie, Magdeburg, Germany, 6Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany, 7 Medizinische Klinik II, Universitätsklinikum Tübingen, Tübingen, Germany, 8 III. Medizinische Klinik, Universitätsmedizin Mannheim, Mannheim, Germany, 9 Klinik I für Innere Medizin, Uniklinik Köln, Köln, Germany 1 2

Introduction: Myelofibrosis (MF) patients (pts) suffer from constitutional symptoms, splenomegaly, and cytopenia. While ruxolitinib (RUX) has been approved to reduce symptoms and splenomegaly, recent data indicate that pomalidomide (POM) may improve anemia. We therefore designed a multicenter phase-Ib/II combination study of RUX plus POM. Methods: Primary endpoints are safety and response rate after 12 treatment cycles (28 days each) according to IWG-MRT criteria or achievement of RBC transfusion independence. Secondary endpoints are quality of life and progression-free / overall survival. Main inclusion criterion is primary or secondary MF with anemia (Hb < 10 g/dL). Key exclusion criteria are suitability for allotransplant and low platelet count (<100/nL). While POM treatment is fixed at 0.5 mg QD, RUX is started at 10 mg BID

Abstracts


CONTENTS AUTHOR INDEX

with dose modifications being allowed. In total, 72 pts are intended to be studied in a Simon´s two-stage design. Results: Here, data from the first 20 pts treated since August 2013 are reported. Regarding safety, 182 adverse events (AE) of any grade (CTCAE °1–5) were recorded. Among these, worsening of anemia was the most frequent AE (n = 63 in 10 pts; °2–4), followed by musculoskeletal pain (n = 13 in 4 pts; °1–2), constitutional symptoms (n = 9 in 2 pts; °1–3), and neutropenia (n = 9 in 1 pt; °4). Most AE were mild and associated with the underlying MF. Treatment interruptions or dose reductions of RUX and/or POM were uncommon. RUX could be step-wise increased to 15 or 20 mg BID in 10/20 pts. Regarding serious AE (SAE), in total 9 events (CTCAE °2–5) was recorded. While 8 SAE (pneumonia °3, n = 2; abdominal pain °5, anemia °4, cardiac decompensation °5, malignant melanoma °2, syncope °3, and TIA °2, n = 1 each) were considered non-related to the study drugs, 1 SAE (neuropathy °3) was likely related to the study medication as it resolved after end of study. Median time on treatment is 7 cycles (range: 2.4–11.9) in 20 pts; 15/20 pts are still on treatment. Five pts discontinued due to death (n = 2), no response after 12 cycles (n = 2) or withdrawal of informed consent (n = 1). Within this short observation time, 3 pts experienced ´Clinical Improvement´ according to IWG-MRT (anemia, n = 2; neutropenia, n = 1). Conclusions: Taking into account the morbidity of the pts, the combination of RUX plus POM is well tolerated. First efficacy data show that in a subset of MF pts hematopoiesis is improved. An update of safety and efficacy will be presented.

the cell and consequently cell polarization. ROCK is a downstream effector of RhoA and leads to stabilization of the actin cytoskeleton and acto-myosin II contraction. The observed reduction of ROCK phosphorylation may reveal an important mechanism of ruxolitinib-induced inhibition of DC migration. Our current efforts focus on the validation of ROCK as off-target JAK1/2-independent target kinase of ruxolitinib as potential mediator of the observed effects, which may at least in part also explain the potential anti-inflammatory therapeutic effects of ruxolitinib.

Disclosure: No conflict of interest disclosed.

Introduction: In eosinophilia-associated myeloproliferative neoplasms (MPN-eo), constitutive activation of protein tyrosine kinases (TK) as consequence of translocations, inversions or insertions and creation of TK fusion genes is recurrently observed. The most commonly involved TKs and their potential TK inhibitors include PDGFRA at 4q12 or PDGFRB at 5q33 (imatinib), FGFR1 at 8p11 (ponatinib) and JAK2 at 9p24 (ruxolitinib). We here report the identification of three new PDGFRB fusion genes in three male MPN-eo patients: MPRIP-PDGFRB in a case with t(5;17)(q33;p11), CPSF6-PDGFRB in a case with t(5;12)(q33;q15), and GOLGB1-PDGFRB in a case with t(3;5)(q13;q33). Methods: Karyotypes were described according to the ISCN 2013. Fluorescence-In-Situ-Hybridization (FISH) was performed with probes which specifically bind the PDGFRB locus 5q33. Analysis of PDGFRB expression by `quantitative reverse transcription polymerase chain reaction´ (qRTPCR) was performed as previously described. The fusion genes were identified by 5´-rapid amplification of cDNA ends polymerase chain reaction (5´-RACE-PCR) or DNA-based long distance inverse PCR (LDI-PCR). Results: Cytogenetic analysis showed reciprocal translocations involving chromosome band 5q33. Subsequent FISH analyses indicated a rearrangement of PDGFRB. The individual PDGFRB/ABL1 ratios were 41 (case #1), 72 (case #2) and 109 (case #3) which were all significantly higher than the level of 23 that was identified previously as the maximal level seen in control individuals. LDI-PCR or 5´-RACE-PCR identified fusion of PDGFRB with MPRIP, CPSF6 and GOLGB1. RT-PCR confirmed an in-frame fusion between MPRIP exon 20 and PDGFRB exon 12 in addition to an in-frame fusion transcript with a deleted MPRIP exon 19 (-119bp) and a truncated exon 20 (-1bp) with PDGFRB exon 12. The fusion transcript CPSF6-PDGFRB revealed an in-frame fusion between CPSF6 exon 5 and PDGFRB exon 11. RT-PCR of GOLGB1-PDGFRB illustrated three different fusion transcripts involving GOLGB1 ex 10 and PDGFRB exons 12, 13 and 14. Conclusion: We have identified three new PDGFRB fusion genes in three male MPN-eo patients. All three patients achieved rapid and durable remissions on imatinib. Our findings underline the major diagnostic role for routine cytogenetic analysis in the diagnostic work-up of eosinophilia.

V723

The JAK inhibitor ruxolitinib impairs dendritic cell migration via off-target inhibition of ROCK activation Rudolph J.1, Heine A.1, Quast T.2, Kolanus W.2, Trebicka J.3, Brossart P.1, Wolf D.1 Department of Internal Medicine III, Oncology, Hematology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany, 2Molecular Immunology & Cell Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany, 3Department of Internal Medicine I, University Clinic Bonn (UKB), Bonn, Germany 1

Introduction: Ruxolitinib is a JAK inhibitor approved for treatment of myelofibrosis and shows impressive symptom control by suppression of inflammation. Ruxolitinib is also a promising drug for treatment of acute and chronic GvHD (Spoerl et al., Blood 2014). The immune-modulatory effects are at least in part due to an inhibitory effect on dendritic cell biology (Heine et al., Blood 2014). Dendritic cells (DCs) are important antigen-presenting cells. Upon antigen contact they migrate into the draining lymph nodes to prime T cells. The aim of this study was to define the impact of JAK inhibition on DC migration. Methods: Migration of human monocyte-derived DCs (moDC) or murine bone marrow-derived DCs (bmDCs) was analyzed in Transwell assays or dynamically by time-lapse microscopy within three dimensional collagen gels towards CCL-19 gradients. Signaling events were analyzed by Western Blot for changes in phosphorylation levels. Results: 2D migration of ruxolitinib-exposed DC is dose-dependently reduced in vitro. By analyzing the migratory phenotype of human moDCs within 3D collagen gels, ruxolitinib-exposed DCs are still able to sense chemokine gradients and form lamellipodia at the leading edge of the cell, whereas the retraction of the uropod is inhibited. Additional in vivo studies could show that the JAK inhibitor potently reduces homing of bmDCs into draining lymph nodes in mice. siRNA knockdown experiments revealed that this inhibitory effect is JAK1- and JAK2-independent. On a molecular level we could show a reduced phosphorylation of Rho-associated protein kinase (ROCK) in ruxolitinib-treated moDC upon CCL-19 stimulation. Finally, the observed migration phenotype could be mimicked by the ROCK inhibitor Y-27632. Conclusions: RhoA family members are key proteins controlling important steps of cell migration, such as protrusion formation at the front of

Abstracts

Disclosure: Janna Rudolph: No conflict of interest disclosed. Dominik Wolf: Financing of Scientific Research: Novartis, BMS, Pfizer, Amgen, AOP; Expert Testimony: Novartis, AOP. V724

Fusion of PDGFRB to MPRIP, CPSF6 and GOLGB1 in three patients with eosinophilia-associated myeloproliferative neoplasms Naumann N.1, Schwaab J.1, Metzgeroth G.1, Jawhar M.1, Haferlach C.2, Schlegelberger B.3, Göhring G.3, Dietz C.T.1, Schnittger S.2, Lotfi S.4, Hofmann W.-K.1, Cross N.C.P.5, Reiter A.1, Fabarius A.1 III. Medizinische Klinik, Universitätsmedizin, Mannheim, Germany, 2MLL Münchner Leukämie Labor, München, Germany, 3Institut für Humangenetik, Medizinische Hochschule Hannover, Hannover, Germany, 4Onkologie MVZ am Siloah St. Trudpert Klinikum, Pforzheim, Germany, 5Wessex Regional Genetics Laboratory, Salisbury, United Kingdom 1

Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

217


CONTENTS AUTHOR INDEX

V725

Treatment with ruxolitinib seems to attenuate the prognostic impact of the phenotype-driver mutations in patients with myelofibrosis Wang S.-Y.1, Jäkel N.1, Krahl R.1, Hubert K.1, Roskos M.2, Niederwieser D.1, Al-Ali H.K.1 University Hospital of Leipzig, Department of Haematology/Oncology, Leipzig, Germany, 2Synlab Jena Oncoscreen, Jena, Germany 1

Introduction: CALR mutation is a favourable prognostic variable in myelofibrosis (MF) compared with JAK2, or MPL mutations. We analysed whether treatment with ruxolitinib (RUX) could modify the prognostic impact of these phenotype-driver mutations in terms of survival in patients (pts) with MF. Patients and methods: An analysis of 127 consecutive patients (pts) with MF at a median age of 58 years seen in our department from Sept. 2009 to May 2014 were included. JAK2V617F, CALR and MPL mutations as well as clinical and cytogenetic risk stratifications were conducted. Results: JAK2 mut+ (group A; 60.6%) constituted the largest group, followed by CALR mut+ (group B; 19.7%), and “triple-negative” (group C; 19.7%). The median duration of MF was 22 months with no difference in the 3 groups. RUX was given to 72 (57%) pts. [group A (66%), group B (56%), group C (29%)] with a median exposure of 8 months. The response of 80% was not influenced by the mutational status, cytogenetics, or IPSS variables. Leukemic transformation (LT) was documented in 22 pts (14 (64%) pts. had no RUX exposure prior LT). Overall, OS at 3-years was 82%, and worst for group C (72%) compared to 80% for group A (p = 0.05) and 96% for goup B (p = 0.003). In univariate analysis, non-mutated status (p = 0.02), Int-2/high-risk IPSS (p = 0.06), anemia (p = 0.03), transfusion dependency (p = 0.04), and platelets < 100 × 109/L (p = 0.06) but not unfavorable cytogenetics were associated with an inferior OS. In multivariate analysis, advanced IPSS only (p = 0.04) but not the mutation status (p = 0.4) retained its negative impact on OS. Although int-2/ high-risk IPSS was more common in group A pts. compared to group B (p = 0.01), OS for group A pts treated with RUX was not statistically inferior to the entire group B cohort. For only int-2/high-risk IPSS pts, OS in group A pts treated with RUX was similar to that of group B pts (p = 0.4). Conclusions: The mutation status in MF bestows distinct clinical phenotypes and has prognostic and therapeutic implications. Response to RUX seems to be independent of the mutation status. More importantly, a JAK1/JAK2 inhibition appears to attenuate the prognostic implication of the different mutation profiles by improving the less-favourable survival associated with non-CALR-mutation status. The full potential of a sustained JAK1/JAK2 inhibition in modifying survival in the context of the various mutational profiles needs to be further studied. Disclosure: Song-Yau Wang: No conflict of interest disclosed. Haifa Kathrin Al-Ali: Financing of Scientific Research: honoraria and research funding.

Freier Vortrag Palliativmedizin V730

Benefit and risk of a percutaneous endoscopic gastrostomy (PEG) for decompression as a palliative intervention in patients with malignant bowel obstruction Dittrich A.1, Schubert B.2, Kramer M.1, Kast K.3, Lenz F.1, Schuler U.1, Schuler M.1,4 Universitätsklinikum Carl Gustav Carus Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany, 2Krankenhaus St. Joseph-Stift, Klinik für Innere Medizin, Dresden, Germany, 3Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Dresden, Germany, 4 HELIOS Klinikum Emil von Behring, Klinik für Innere Medizin II, Berlin, Germany 1

Introduction: Nausea and vomiting due to malignant bowel obstruction (MBO) present many burdens for patients suffering from end-stage abdominal cancer. Percutaneous endoscopic gastrostomy (PEG) has been proposed as a therapeutic option for decompression. This retrospective study investigates whether there is a relevant decrease in symptoms due to PEG and the corresponding complications in patients of two palliative care departments in Dresden, Germany. Methods: Chart reviews of 75 patients who underwent PEG tube placement for decompression between 2002 and 2013 were performed. Abstracted data includes demographics, symptoms (vomiting, nausea, abdominal pain) and use of antiemetics and analgetics up to 7 days before and after intervention, use and acceptance of a nasogastric tube (NG tube), potential risk, prognostic and influencing factors, complications, and survival. A linear mixed model with random intercept and generalized estimation equations (GEE) were used to determine symptom reduction considering the influence of medication. Results: The mean frequency of vomiting per day was reduced from 2.0 (95% CI: 1.7–2.4) without any therapy to 0.1 (95% CI: 0.0–0.7) with PEG (p < 0.001). The probability of the occurrence of nausea on a given day was 59% (95% CI: 47–70%) prior to the PEG placement and 27% (95% CI: 13–48%) afterwards (p < 0.001). Before the PEG, 53 patients (71%) temporarily had a NG tube for symptom relief, which led to a decrease in vomiting from 2.0 to 0.4 (95%CI: 0.0–1.1) times a day (p < 0.001). 80% of these patients reported poor toleration of the NG tube. 112 complications were reported in 56 patients (none: 19 patients (25%), minor: 42 (56%), moderate: 27 (36%), severe: 9 (12%)). The median overall survival after the procedure was 28 days (range 2–440). 59% of the patients could be discharged after a median postprocedural stay of 9 days (range 1–28). The readmission rate due to complications or recurrent symptoms was 12%. Conclusion: The PEG placement leads to a significant reduction of vomiting and nausea in patients with MBO. Minor complications such as leakage, occlusion and mild wound pain occur quite often and should be discussed with the patient prior to intervention. Nevertheless, it seems that benefits prevail over risks, with severe complications being rare. Since surgical and nonsurgical attempts of alleviating ileus symptoms in patients with MBO often fail, one should consider placing the PEG earlier in the course of the disease. Disclosure: No conflict of interest disclosed.

218

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V731

Sequential assessment of ouality of life, distress, depression, anxiety and symptom burden in patients with advanced cancer – a pilot study Weber S.1,2, Gerlach C.1,2, Hopprich A.1,2,3, Reinholz U.1,2, Hess G.2,3, Theobald M.2,3, Weber M.1,2 University Medical Center of the Johannes Gutenberg-University of Mainz, Interdisciplinary Palliative Care Unit, III. Department of Medicine, Mainz, Germany, 2University Medical Center of the Johannes Gutenberg-University of Mainz, University Cancer Center (UCT Mainz), Mainz, Germany, 3University Medical Center of the Johannes Gutenberg-University of Mainz, III. Department of Medicine, Mainz, Germany 1

Introduction: Patient-reported outcome measurement (PROM) might be a way to identify patients with high symptom burden for early referral to palliative care. The primary aim of this study is to evaluate the feasibility of providing PROM on several aspects of quality of life (QoL) in patients with advanced cancer in a German academic out-patients clinic at intervals of 6 weeks. Methods: Eligible patients had an estimated clinical prognosis of 3–12 months as assessed by the Surprise-Question (“Would I be surprised if this patient died within the next year?”), advanced cancer, ECOG PS 0–2 and no cognitive impairment. Quality of life (FACT-G), symptom burden (MIDOS), depression and anxiety (PHQ-4) as well as distress (NCCN-Distress Thermometer) were measured at baseline and after 6, 12, 18 and 24 weeks. The primary outcome was the evaluation of feasibility of providing PROM for ambulatory patients with advanced cancer. Secondary endpoints included the results of PROM for the mentioned QoL aspects. Results: 1153 patients were screened within 4 months, 129 patients assessed for eligibility and 42 patients included. 19 patients completed 5 surveys (number of surveys available for analysis, see fig. 1). Reasons for missed surveys were: died within the course (N = 10), declined to participate (N = 3), administrative failure (N = 11) (see fig.1). 160 surveys were available for analysis. 54% of the surveys documented a clinically significant distress (DT score ≥5). 19% of 160 surveys documented moderate and 11% severe pain. Moderate and severe breathlessness were indicated by 23% respectively 6%. 33% of the surveys documented moderate and 16% severe weakness, related to that, 34% of the surveys indicated moderate and 14% severe tiredness. 14% surveys with a PHQ-4 Score ≥6 suggested the presence of a depressive or anxiety disorder.

Conclusions: PROM is a feasible method to identify patients with high symptom burden at an outpatient oncology clinic. Patients with advanced cancer are willing to participate in PROM. Patient, disease and staff-associated barriers to providing PROM need further research for a useful implementation in oncology practice. Disclosure: No conflict of interest disclosed. V732

Results from a time and motion study of denosumab subcutaneous injection and zoledronic acid intravenous infusion in patients with metastatic bone disease from German sites Wimberger P.1, Sommer U.2, Nietzke M.2, Gatta F.3, Ikenberg R.4, De Cock E.5, Tao S.6, Kritikou P.7, Body J.-J.8, Hechmati G.3 University Hospital Carl Gustav Carus, Dresden, Germany, 2St. Johannes Hospital, Dortmund, Germany, 3Amgen (Europe) GmbH, Zug, Switzerland, 4 Amgen GmbH, Munich, Germany, 5United BioSource Corporation, Barcelona, Spain, 6United BioSource Corporation, Montreal, Canada, 7United BioSource Corporation, London, United Kingdom, 8CHU Brugmann, ULB, Brussels, Belgium 1

Introduction: In cancer patients with metastatic bone disease, the mainstay treatment for skeletal-related events prevention was zoledronic acid (Zol) administered as an intravenous (IV) infusion. Since 2011, denosumab (Dmab) subcutaneous (SC) injection is a superior treatment option providing an alternative formulation to IV infusions. This Time and Motion study was conducted to estimate time of all activities associated with Dmab SC injection and Zol IV infusion monotherapy. Methods: In the outpatient setting of 2 German hospitals, patients with bone metastases from solid tumours were observed during administration of Dmab SC or Zol IV. The following pre-defined tasks were recorded in military time: patient registration (Site 01: it additionally includes blood test and vital signs check), IV catheter installation, IV/SC drug administration (IV: infusion connection until disconnection; SC: syringe filling and injection), and post-treatment monitoring. The time that a healthcare professional (HCP) actively dedicated to perform a pre-defined task was recorded using a stopwatch. Time endpoints measured for each treatment were total task time, total active HCP time, drug administration duration, patient chair time and patient time in care unit. Data (mean) collected per site were analysed by treatment and pooled across sites. Results: Table shows data recorded per site from a total of 80 observations as well as the pooled results. The pooled analysis of mean administration duration of Dmab SC injection was 2.7 minutes (min) which was nearly 10 times faster than Zol IV infusion (26 min). The pooled mean total task time was 8.5 min for Dmab SC and 51.8 min for Zol IV (–84%). Time savings associated with Dmab SC were also reflected in the total active HCP time and patient times in the chair and care unit, with reductions of 44%, 85%, and 48%, respectively. Conclusions: Use of Dmab SC injection was associated with substantial time savings for all outcome measures. Opting for Dmab SC administration instead of Zol IV infusion should decrease HCP time and increase the outpatient capacity to treat more patients in Germany.

Fig. 1. STROBE-Diagram.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

219


CONTENTS AUTHOR INDEX

Tab. 1. Data collected per site and pooled results

Site 01

Site 02

Pooled Data

Mean time in minutes

Dmab SC (n = 20)

Zol IV (n = 20)

Dmab SC (n = 20)

Zol IV (n = 20)

Dmab SC (n = 40)

Zol IV (n = 40)

Drug Administration Duration

2.7

39.3

2.7

12.8

2.7

26.0

Total Task Time

11.5

68.8

5.5

34.9

8.5

51.8

Total Active HCP Time

9.1

13.7

3.3

8.5

6.2

11.1

Patient Chair Time

12.8

74.4

3.3

33.2

8.0

53.8

Care Unit Time

72.2

123.5

15.8

45.7

44.0

84.6

Disclosure: No conflict of interest disclosed.

Disclosure: Pauline Wimberger: Financing of Scientific Research: Amgen, Roche, Novartis, Merck, TEVA and AstraZeneca. Guy Hechmati: Employment or Leadership Position: Amgen; Stock Ownership: Amgen. V733

Symptoms and needs of head and neck cancer patients at diagnosis of incurability – an APM prospective longitudinal multicenter cohort study Seidel W.1, Lordick F.2, Vogt J.2, Mehnert A.3, Thomas M.4, van Oorschot B.5, Wolff H.6, Schliephake H.7, Canis M.8, Nauck F.1, Alt-Epping B.1 Universitätsmedizin Göttingen, Klinik für Palliativmedizin, Göttingen, Germany, 2Universitäres Krebszentrum Leipzig (UCCL), Leipzig, Germany, 3 Universitätsklinikum Leipzig, Abteilung für Medizinische Psychologie und Medizinische Soziologie, Leipzig, Germany, 4Thoraxklinik Heidelberg, Heidelberg, Germany, 5Universitätsklinikum Würzburg, Interdisziplinäres Zentrum für Palliativmedizin Klinik für Strahlentherapie, Würzburg, Germany, 6Universitätsmedizin Göttingen, Klinik für Strahlentherapie und Radioonkologie, Göttingen, Germany, 7Universitätsmedizin Göttingen, Klinik für Mund-Kiefer-Gesichtschirurgie, Göttingen, Germany, 8Universitätsmedizin Göttingen, Klinik für Hals-Nasen-Ohrenheilkunde, Göttingen, Germany 1

Introduction: Although substantial efforts have been made to better understand palliative care needs of patients (pts.) suffering from very advanced stages of cancer near the end of life, very little is known about physical symptoms and the psychosocial burden of pts. at the time of diagnosing an incurable situation. This, though, would be even more necessary, as S3 cancer guidelines as well as modern palliative care concepts demand early integration of palliative care concepts, beginning from the diagnosis of incurability. Methods: Therefore, the Arbeitsgemeinschaft Palliativmedizin (APM) of the German Cancer Society (DKG) initiated a prospective longitudinal multicenter cohort study, assessing pts. suffering from incurable cancer with respect to symptom burden and psychosocial needs, from the time of diagnosing incurability (i.e., before palliative anticancer treatment was initiated) and in three-monthly intervals thereafter, using FACT, SEIQoL-Q, PHQ-4, a modified SCNS-SF-34, and the NCCN Distress Thermometer. Here, we present preliminary results of the Göttingen study site, with a distinct focus on pts. suffering from head and neck squamous cell cancer (HNSCC); incurability was defined as inoperable local primary tumor or relapse, or metastatic disease. Results: Until 04/2015, 10 pts. gave formal consent and were included into the study (9 pts. with HNSCC, 1 pt. with SCC of the scalp, infiltrating the neck and parotid gland). 6 pts. were male, 4 female; mean age was 62.4 years (median 59 years). ECOG scores were almost equally distributed between 0 (2 pts.) and 4 (2 pts.) at first visit. Pts. described a broad spectrum of physical symptoms and psychosocial needs, with strong variations in intensity from virtually no feeling of being compromised to extremely

220

perceived suffering (mean value of the 0–10 NCCN distress scale: 7,5). The subjective relevance of the inability to eat what was desired and of xerostomia, and the importance of family support were found to be repetitive patterns. Conclusions: Even at first visit / at the beginning of an incurable course of disease, a broad spectrum of needs and symptoms were identified, even before palliative anticancer treatment was initiated. These preliminary findings underline the need for early, pro-active symptom and distress screening and for the individualized provision of multifaceted support, including palliative care services, beginning even from the time of diagnosis of incurability.

Oncol Res Treat 2015;38(suppl 5):1–270

V734

Acute Myeloid Leukaemia patients in the palliative care setting: Analysis of life expectancy, quality of life and transfusion frequency Kriesen U.1, Riedel J.1, Große-Thie C.1, Leithäuser M.1, Gläser D.1, Gläser H.1, Junghanß C.1 Universitätsmedizin Rostock, Zentrum für Innere Medizin, Klinik III, Hämatologie/Onkologie, Palliativmedizin, Rostock, Germany 1

Introduction: The prognosis of Acute Myeloid Leukaemia (AML) especially in elderly patients is grave. As intensive chemotherapy courses aiming at the cure of the patients often fail in these patients it is of particular importance to provide them a structured palliative care. Symptom load of AML palliative care patients include anaemia, bleedings as well as coping strategies. Here we present data on 35 AML patients which were treated in the palliative care ward of the University of Rostock during recent years. Methods: We performed a retrospective analysis of all AML patients which were referred to the palliative care ward of the University of Rostock from August 2008 to October 2014 (n = 35). Analysis included patients´ characteristics like age, AML characteristics as well as transfusion frequency in regard to hemoglobin as well as thrombocyte levels and the clinical benefit. Results: AML patients’ age ranged from 54–89 years. Our current results indicate that red blood cell transfusions were given at mean hemoglobin level of 4.9 mmol/l (mean 3.3 units/ patient). Patients hemoglobin level increased thereafter to a mean 6.1 mmol/l and reported clinical benefit in regards to dyspnea, weakness and tiredness. Mean platelet counts at transfusion were 11.0 GPT/l. Patients received the mean of 3.2 platelet units. Visible bleedings could be reduced. Common treatment approaches were physiotherapy aiming at higher mobility, improving of the coping by the palliative care stuff including psycho-oncologists as well as advanced care planning in regards to discharge and patient will. Conclusion: This early retrospective study demonstrated that transfusions are frequently performed in AML patients which are referred a palliative care ward. This issue often precludes patients from transfer to a hospice institution. Indications for a transfusion have to be discussed with the patients in detail, however transfusions are still performed frequently leading to a good symptom control. Further prospective studies are necessary to address these patients in the palliative care setting. Disclosure: No conflict of interest disclosed. V735

Transfusions in the last 30 days before death of terminal ill patients in palliative care – time of survival and quality of life Kriesen U.1, Römer S.1, Leithäuser M.1, Gläser D.1, Gläser H.1, Große-Thie C.1, Junghanß C.1 Universitätsmedizin Rostock, Zentrum für Innere Medizin, Klinik III, Hämatologie/Onkologie, Palliativmedizin, Rostock, Germany 1

Introduction: Blood- and platelet transfusions are of particular importance in supportive treatment of anemia associated with impairing symptom burden in palliative care. Mainly driven by the idea of enabling a

Abstracts


CONTENTS AUTHOR INDEX

dying process with a minimum amount of symptoms, this study is about to figure out the extent of benefit from transfusions and the effect to the premortal quality of life in palliative care. Methods: The retrospective study is based on patient data of the palliative care unit of Rostock University from 2013 and 2014. We thereby analyze data of two groups. First group was defined by deceased patients that where given blood and platelet transfusions within the last 30 days of life (n = 34). The reference group was set to the same amount of patients, without transfusion therapy premortal. To examine the specific effect of transfusions on quality of life, following parameters where chosen to match the two patient groups: age, gender and ECOG performance status on hospital admission. The current study will provide insight into shift of personal stress during the patient’s last hospitalization. The stress level will be identified on parameters such as dyspnea, weakness, fatigue and fear. The impact of the parameters is given by its amplitude on the numeric rating scale (NRS). Furthermore objective parameters such as level of hemoglobin, count of erythrocytes and thrombocytes, blood clotting and time of survival, hereby defined as the time between the admission of last hospitalization period and death, will be used to support robust results. Conclusion: Based on the study “Use of Blood Transfusion at the End of Life” by Goksu et al from 2014 we expect that transfusions at the end of life do have significant positive impact on survival time of terminally ill patients. Furthermore we expect that transfusions do have positive impact on personal symptom burden. However, it has to be taken into consideration that an expansion of survival not only lengthens life but also possibly prolongs ailment. Based on that, it is of major importance to balance the benefit in symptom burden against associated stress and alternative options in treatment. Therefore the individual situation is crucial for the treatment method to be chosen. Finally, consent or refusal of the dying patient is the most import criterion for the decision in end of life care. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Supportive Therapie II V742

Impact of physical exercise on upper body dysfunction and fatigue in breast cancer patients during radiotherapy Wehrle A.1, Ilaender A.2, Kneis S.3, Volegova-Neher N.4, Scholber J.4, Bertz H.3 Universitätsklinik Freiburg, Institut für Bewegungs- und Arbeitsmedizin, Freiburg, Germany, 2Klinik für Tumorbiologie, Freiburg, Germany, 3 Universitätsklinik Freiburg, Klinik für Innere Medizin I, Schwerpunkt Hämatologie, Onkologie und Stammzelltransplantation, Freiburg, Germany, 4 Universitätsklinik Freiburg, Klinik für Strahlenheilkunde, Freiburg, Germany 1

Introduction: Breast cancer patients often suffer from different side effects of their disease and therapy (surgery/chemo-/radiotherapy). Besides specific side effects like upper body dysfunction with limited shoulder range of motion of the affected limb, many patients show a general decrease in physical and psychosocial capacity. In sum these aspects result in a decreased quality of life. Even though there are publications showing a beneficial effect of physical activity for breast cancer patients, already during treatment, only few studies investigated exercise during radiotherapy. The aim of this study was to evaluate whether an exercise intervention may reduce aforementioned impairments. Furthermore feasibility of handheld vibration training and the effects on upper body function should be tested. Methods: 21 female breast cancer patients (age: 58 ± 12.36 years) planned for radiotherapy were allocated either to an intervention group (IG) or a passive control group (CG) as they preferred. IG performed an exercise program (3x /week) during six weeks of radiotherapy including warm up on a bicycle ergometer, handheld vibration and balance training. Following assessments were performed pre and post radiotherapy or interven-

Abstracts

tion (T0, T1), respectively: Shoulder mobility (range of motion), balance control (sway path of centre of force), maximum lower body performance (jump height), cardiorespiratory fitness (Vo2peak, Pmax), quality of life (EORTC-QLQ C30) and fatigue (MFI). Results: After intervention IG showed improved shoulder mobility in terms of significantly better abduction (P = 0.012) and external rotation (P = 0.026). General fatigue (P = 0.49) as well as mental fatigue (P = 0.023) of IG were significantly reduced, whereas quality of life could not be influenced by the intervention. Furthermore, results of Pmax (P = 0.016) and jump height (P = 0.020) showed a group x time effect, while our intervention did not affect balance control or cardiorespiratory fitness (Vo2peak). Conclusion: The feasibility of handheld vibration training during radiotherapy could be confirmed. Benefits were especially detected for shoulder mobility that was triggered by vibration exercise. In general, our intervention could positively influence fatigue and physical capacity. Therefore our results are in line with previous publications, but are limited by lack of randomization. Disclosure: No conflict of interest disclosed. V743

The impact of NFAT inhibition in neutrophil antifungal defense in a murine Aspergillus fumigatus pneumonia model Teschner D.1, Michel C.1, Prüfer S.2, Theobald M.1, Schild H.2, Radsak M.1,2 University Medical Center of the Johannes Gutenberg-University, Department of Hematology, Medical Oncology, & Pneumology, Mainz, Germany, 2University Medical Center of the Johannes Gutenberg-University, Institute of Immunology, Mainz, Germany 1

Introduction: Opportunistic infections with Aspergillus fumigatus (A. f.) are a major threat to immunodeficient patients after allogeneic stem cell transplantation (HSCT), partly due to immunosuppressive medication e.g. by calcineurin inhibitors like cyclosporine A (CsA). The nuclear factor of activated T cells (NFAT) is known as an important transcription factor downstream of calcineurin in the adaptive immune systems, but plays an important role in innate immune response by polymorphonuclear neutrophils (PMN), as indicated by recent data in rodent models. Methods: We used a murine A. f. pneumonia model (C57BL/6) to study the antifungal innate immune response in vivo under NFAT inhibition. Therefore, we first treated mice intraperitoneally with CsA (18mg/kg/d) or vehicle for 2 weeks and challenged them with A. f. conidia intratracheally. 24 hours later, some mice were sacrificed and PMN recruitment to the lungs and pulmonary fungal clearance were examined. In addition, survival of remaining infected mice was analyzed with neutropenic animals (by depletion with anti-Gr1) serving as positive controls. Secondly, LysM-specific NFATc1 knockout (NFATc1LysM) mice were bred lacking NFATc1 expression solely in myelomonocytic cells (i. e. PMN and monocytes). Furthermore, these animals were infected with A. f. and analyzed as described above. Results: While the infection was lethal in CsA or vehicle treated neutropenic mice, all CsA or vehicle treated mice survived the infection. CsA treated mice showed enhanced PMN recruitment in BAL (55.2% ± 12.0, vs. 33.7% ± 8.0, mean ± SEM), whereas pulmonary inflammation was comparable to controls. In contrast, fungal clearance was impaired in animals after CsA treatment (2.1 × 105 ± 0.5 CFU/lung vs. 1.7 × 105 ± 0.2). Along with that, NFATc1LysM mice infected with A. f. showed unimpaired survival. However, there were no detectable differences in PMN recruitment or fungal clearance, whereas pulmonary inflammation seemed to be more pronounced in knockout mice (1.7 inflammation points ± 0.2 vs. 0.8 ± 0.2). Conclusion: In a mouse model, NFAT inhibition via treatment with CsA does not impair survival after infection with A. f. in vivo but seems to affect PMN recruitment and local fungal clearance. However, infected NFATc1LysM mice displayed enhanced pulmonary inflammation compared to controls. Further studies are needed to clarify underlying mechanisms and clinical relevance in HSCT of our findings. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

221


CONTENTS AUTHOR INDEX

V744

Respiratory virus infections in hematologic and immunocompromised patients: Clinical presentation, diagnosis, treatment and outcome Dörfel D.1,2, Beck R.3, Hofer S.1, Kanz L.1, Vogel W.1, Haen S.P.1,4 Medizinische Universitätsklinik Tübingen, Abteilung II für Onkologie, Hämatologie, Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Department for Internal Medicine II, Eberhard Karls University, Tübingen, Germany, 3Institut für Medizinische Virologie und Epidemiologie der Viruskrankheiten, Tübingen, Germany, 4Interfakultäres Institut für Zellbiologie, Abteilung Immunologie, Tübingen, Germany 1

Introduction: Respiratory virus infections can represent a serious challenge in immunocompromised patients. Without surveillance of the immune system, effective virus clearance is interminable and treatment has to be of long duration. Moreover, antiviral medication might even deteriorate immunosuppression through additional myelotoxicity. In this study, we evaluated the clinical presentation, treatment and prognosis of respiratory virus infections (HMPV, RSV, Influenza) in immunocompromised patients. Methods: We retrospectively analyzed patient records from hematologic patients and/or patients with immunosuppression treated at our institution between 2011 and 2014 and identified 139 patients (67 women, 72 men; median age at detection of virus 59 years, range 17–85 years) with respiratory virus infections (HMPV n = 13, RSV n = 29, Influenza n = 97). Patients suffered from acute (ALL n = 5; AML n = 16) or chronic (CLL n = 7; CML n = 2) leukemia or T-LGL (n = 1), Hodgkin’s (n = 3) or non-Hodgkin lymphoma (n = 11), multiple myeloma (n = 15), myelodysplastic (n = 4) or myeloproliferative syndromes (n = 6), solid tumors (n = 6), or sarcoidosis (n = 3) or received immunosuppressive medication due to chronic lung (n = 46) or rheumatologic diseases (n = 16). 37 patients had a history of allogeneic hematopoietic cell transplantation. Results: Patients presented with couch and dyspnoe (n = 74) and fever (n = 112). Respective viruses were detected in respiratory tract material (smears, BAL). HMPV was treated with Ribavirin (n = 4), Aciclovir (n = 6) and Valcyte (n = 1) or with only symptomatic medication (n = 2). Patients with RSV were treated with Ribavirin (n = 16) or with symptomatic medication (n = 13). Influenza was treated with Oseltamivir (n = 79) or symptomatically (n = 18). Overall virus-related mortality for HMPV, RSV and Influenza patients was 0% (0/13), 20% (6/29) and 7% (7/97), respectively. In the RSV group, 4/16 patients (25%) died despite treatment, and 2/13 patients died after symptomatic therapy (15%). In the influenza group, all patients dying from virus infection were treated with Oseltamivir. Conclusions: Respiratory virus infections in immunocompromised patients, especially with RSV can represent a therapeutic challenge and are associated with high mortality even if patients are treated early with antiviral drugs. Disclosure: No conflict of interest disclosed. V745

Assessing the burden of oral mucositis Berger K.1, Bollig A.1, Strobach D.2, Rieger C.1, Ostermann H.1 Klinikum der Universität München, Medizinische Klinik und Poliklinik III, München, Germany, 2Klinikum der Universität München, Apotheke, München, Germany 1

Introduction: Oral mucositis (OM) is a frequent side effect following anti-cancer therapy. It affects treatment outcome, is related to complications like fever or infections, increases resource use and influences quality of life (QoL) negatively. Despite this impact on cancer treatment only few data on the burden of mucositis have been published so far. Objective of this study was to assess the burden of mucositis in haemato-oncology patients.

222

Oncol Res Treat 2015;38(suppl 5):1–270

Methods: 1. Structured PubMED literature research using MESH terms. Literature evaluation according to health technology criteria (GRADE). 2. Retrospective observational mono-centre feasibility study on treatment patterns, resource consumption of patients (pts) at high risk of developing OM due to high dose methotrexate (MTX) treatment. Data sources: clinic’s electronic data capturing system and patient charts. 3. Questioning of health-care staff on resource use. Results: 1. Screening of a total of 2249 hits yielded 56 original articles describing OM, only very few publications cover German data. Limited German epidemiological data depict incidence ranging from from 20% to 53% for stem cell transplant pts (OM WHO grade 3–4). Some studies show a negative OM impact on QoL, no German study could be identified. Costs associated to OM ranges from Euro 50 up to Euro 2,000 per OM episode. 2. Data from 15 patients (8 lymphoma pts, 7 ALL pts) with high dose MTX chemotherapy was documented. 8 pts were female, average age was 61 yrs (35–78yrs). OM was documented in only 7 pts. Information on OM risk factors like smoking, alcohol consumption was missing in about 80%. OM grading and documentation was not homogenous. Pain due to OM was reported in only 5 pts. Retrospective data did not allow a correlation between OM and side effects like infection, fever.3. Health care professional estimated time for training of OM prophylaxis was 5–10 minutes. Support for daily mouth hygiene measures in pts with grade 3 and 4 OM was 20 minutes and 20 minutes for help with food intake. Conclusions: German publications on OM are limited. There is a need for more comprehensive data on OM. In our center OM seems to be severely underreported. Structured assessment and grading of OM has to be improved. Caring for pts with OM is time intensive, increased resource use has so far no impact on staffing and reimbursement. Prospective observational studies in routine care using OM specific data collection tools are required to determine the burden of OM. Disclosure: No conflict of interest disclosed. V746

Gait analysis in patients after alloHCT Pahl A.1, Straub E.2, Walz I.2, Kneis S.1, Ilaender A.-K.3, Wehrle A.4, Mumm A.3, Bertz H.1 Universitätsklinikum, Klinik für Innere Medizin I, Hämatologie, Onkologie und Stammzeltransplantation, Freiburg, Germany, 2Institut für Sport und Sportwissenschaft, Universität, Freiburg, Germany, 3Klinik für Tumorbiologie, Freiburg, Germany, 4Institut für Bewegungs- und Arbeitsmedizin, Department Innere Medizin, Universitätsklinikum, Freiburg, Germany 1

Introduction: Continuous and intensive medical treatment often causes postural instability, which may lead to gait disturbances and thus an increased risk of falling. Additionally, gait speed is associated with both, mortality and morbidity. The most intensive medical treatment in oncology is an allogenic hematopoietic cell transplantation (alloHCT), which entails the onset of numerous side effects including decreased physical functioning. However, studies analyzing dynamic postural control in this context are still lacking. Therefore the aim of this study is to investigate gait stability in patients after alloHCT (Pat) using sensor soles (OpenGo science ®) and compare them to healthy matched controls (Con) and seniors (Sen). Methods: A total of 49 participants were included in this cross sectional study (Pat: n = 17, mean = 55 ± 11,12 years; Con: n = 15, mean = 55 ± 10,13 years; Sen: n = 17, mean = 77 ± 3,32 years). Over a distance of 10 meters we measured the number of heel strikes (n), gait speed (km/h), cadence (steps/min), double support time (s) and stride-to-stride variability (%) under three different conditions: normal, fast and dual task walking. Results: Normal walking did not result in any group differences. In the fast walking condition Pat demonstrated a significantly lower gait speed (p < 0.01), lower cadence (p < 0.01), longer double support time (p = 0.01) and a higher number of heel strikes (p < 0.05) compared to Con. During dual task walking significant differences between groups could only be detected regarding gait speed performance (p < 0.05). Moreover, the increase in gait speed comparing normal to fast walking was significantly

Abstracts


CONTENTS AUTHOR INDEX

higher in Con than in Pat (C: +66%, P: +40%, p < 0.05). Additionally Pat >60 years demonstrated a lower gait speed (–21%), than Pat <60 years . A comparison of Sen with Pat and Con shows no significant different in any walking condition. Conclusion: Pats’ poor performance in comparison to Con, especially the inability to increase gait speed, is in line with our hypothesis that patients after alloHCT suffer from impairments of mobility and physical functioning. Although the comparison of Sen with Con and Pat did not render significant results, a consequent sub-group analysis indicated that seniors after alloHCT may be particularly at risk of falling and morbidity. Therefore physical exercise interventions should be implemented early on to reduce the loss of physical functioning and improve the rehabilitation process. Disclosure: No conflict of interest disclosed. V747

Nintendo Wii® versus physiotherapeutic treatment in the setting of hematopoietic stem cell transplantation Schumacher H.1, Strüwe S.1, Greger N.1, Blachke P.1, Freitag S.1, Junghanss C.1, Hilgendorf I.1,2 Universitätsmedizin Rostock, Klinik für Hämatologie, Onkologie und Palliativmedizin, Rostock, Germany, 2Universitätsklinikum Jena, Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistiche Onkologie, Jena, Germany 1

Introduction: The positive effects of physical and sports therapy for strain dependent physical practice are well known. Nevertheless, the available capacities and problem orientated therapies during stationary treatment and aftercare operations, in patients receiving hematopoietic stem cell transplantation (HSCT) are limited. Methods: Here we report on a prospective, randomized study comparing a multimedia sensor-based practice with a classical physiotherapeutic treatment in 49 HSCT recipients. Both groups performed the exercises under the supervision of a physiotherapist. Physical function was evaluated at the date of hospital admission (T1), 14 days (T2) and 30 days after HSCT (T3). Conditional status was assessed by Treadmill and a two minute walk test (2-MWT). Furthermore, a grip strength test was performed with a hydraulic hand dynamometer and balance was evaluated using the Berg Balance Scale. Results: HSCT-recipients were randomized into the control group receiving physiotherapeutic treatment (n = 30) or the experimental group with exercising on the Nintendo Wii® (n = 19). The median age of patients was 59 (43–69) years in the control group and 56 (21–65) years in the experimental group. Treadmill intervention showed a mean performance at T1/T2/T3 of 83 (25–125) W / 68 (25–125) W / 69 (25–125) W in the control group and 104 (75–150) W / 82 (50–125) W / 92 (50–125) W in the Nintendo Wii®-group. In the 2-MWT the median distances at T1/T2/ T3 in the control group were 149 (100–211) / 144 (75–202) / 154 (100– 205) m, while the experimental group walked an average distance of 163 (135–195) / 150 (60–208) / 145 m (80–218), respectively. The grip strength test resulted in mean values of 31 kg (T1), 27 kg (T2 and T3) in the control group and 38 kg (T1), 35 kg (T2), 37 kg (T3) in the experimental group. The control group scored at T1/T2/T3 a mean value of 55/54/54 in Berg Balance Scale, compared with 55/55/55 in the experimental arm, respectively. Conclusion: The functional status of patients decreased after HSCT in both groups and impairment remained at least until one month after HSCT. However, compared to classical physiotherapeutic treatment the group with the sensor-based practise showed a slightly lower endurance, while none differences was observed in balance testing. Exergaming is feasible as well as enjoyable in the stationary setting of HSCT and may be used in addition to physiotherapeutic treatment. Disclosure: No conflict of interest disclosed.

Abstracts

Freier Vortrag

Versorgungsforschung V748

Follow-up care of breast cancer patients who were treated in a German breast cancer centre – survey of patients and attending physicians and analysis of treatment data Feiten S.1, Dünnebacke J.2, Friesenhahn V.1, Heymanns J.3, Köppler H.3, Meister R.1, van Roye C.3, Thomalla J.3, Wey D.2, Weide R.3 Institut für Versorgungsforschung in der Onkologie, Koblenz, Germany, Katholisches Klinikum Koblenz-Montabaur, Brustzentrum im Marienhof, Koblenz, Germany, 3Praxisklinik für Hämatologie und Onkologie, Koblenz, Germany 1 2

Introduction: Breast cancer treatment leads to long-lasting impairments which, according to international guidelines, have to be identified and treated in follow-up care. It remains unclear how follow-up care is perceived by patients and if all needs are met in routine care. Methods: All women who underwent surgery in a German breast cancer centre from 2007 to 2013 were asked to fill out a standardized scanner-readable questionnaire. Medical data were retrieved from their charts and statistically analyzed together with the questionnaire responses. Physicians who could possibly care for breast cancer patients after primary therapy, such as gynecologists, oncologists, radiologists, general practitioners or specialists in internal medicine were invited to fill out a standardized scanner-readable questionnaire as well. Results: 920 questionnaires were filled out and returned (response rate: 61%) by patients. Median age at the time of the survey was 65 years (32– 95). 58% of patients still received some form of therapy, 95% of them hormonal therapy. 94% were still in follow-up care, 5% stopped and 1% never went. Intervals of follow-up visits suggested by international guidelines were assessed as “quite right” in 93%. The following examinations were conducted throughout the whole follow-up period at least once: physical examination (93%), mammography (90%), sonography of breast (81%) and liver (22%), laboratory (56%), tumor marker (23%), MRI (20%) and CT (15%). Different items were rated on a 6-point scale ranging from “0” “not true at all” to “5” “completely true”. Follow-up care was regarded as very important for the own health (4.7), reassuring and calming (4.5), well-being to be looked after (4.4) and well cared for (4.4). A continuous contact between patient and doctor was appreciated (4.4). Visits were connected only to a part with distress (2.1), the median score on the NCCN distress thermometer was 4 (0–10). Analyses of treatment data and the survey of follow-up care specialists will be presented at the meeting. Conclusions: An overwhelming majority of patients makes use of follow-up care. Most important qualities from the patient´s perspective are reassurance, a feeling of security, calming and continuous care by their doctor. Examinations which are not recommended in international guidelines are used in a minority of patients only. Disclosure: No conflict of interest disclosed. V749

Perceptions of non-physician staff about oncologists in their role as managing directors Osburg S.1, Hermes-Moll K.1, Walawgo T.1, Baumann W.1 Wissenschaftliches Institut der Niedergelassenen Hämatologen und Onkologen (WINHO), Köln, Germany 1

Introduction: High staff fluctuation is an issue in many oncology practices. The relationship to the management can be seen as an indicator to optimize continuous employment. Thus, WINHO conducted a survey questioning non-medical staff about their perceptions regarding social support received from the management focusing on good conversational atmosphere, assistance in difficult work situations as well as praise and appreciation for their work effort. Results on the social support from the

Oncol Res Treat 2015;38(suppl 5):1–270

223


CONTENTS AUTHOR INDEX

management are presented and compared from two surveys carried out in 2011 and 2013. Methods and Materials: The study based mainly on the questionnaire “MIKE” developed by the Institute for Medical Sociology, Health Services Research and Rehabilitation Science of the University of Cologne containing question items with Likert scales from 4 to 7. In 2013 and 2011, all 450 hematologists and oncologists from the WINHO network were invited by mail to participate in the survey. 56 specialists from 21 oncology practices participated in both years. Results: In both samples, almost all of the interviewed employees were females (over 90%) working in therapy assistance, reception and laboratory. In 2013, the study population consisted of 434 non-physician staff and in 2011 of 570 employees. Focused on the social support from the management the results are: conversational atmosphere (2011: 78.90%, 2013: 80.72), assistance in difficult work situations (2011: 81.23%; 2013: 84.12) and praise and appreciation for work effort (2011: 70.65%, 2013: 75.76%). In 2013, better outcomes were reached in all categories. The differences between 2011 and 2013 are significant regarding assistance in difficult work situations and praise and appreciation for work effort (Mann-Whitney-U-Test: 0.016 and 0.000). Conclusions: It seems that the oncologists have improved their behavior towards non-physician staff through support in difficult work situations and also through praise and appreciation. Hence, there is evidence that hematologists and oncologists work with the results of the survey in order to improve the situation of the non-physician staff. By contrast, there are specialists who did not participate in the second round. More measures, which are used to enhance the relationship to the staff, will be presented on the congress. Disclosure: No conflict of interest disclosed. V750

Measuring of process quality with indicators – a critical view based on results of a pilot study Hermes-Moll K.1, Hauer A.1, Klein A.1, Baumann W.1, Schmitz S.2 Wissenschaftliches Institut der Niedergelassenen Hämatologen und Onkologen (WINHO) GmbH, Köln, Germany, 2Gemeinschaftspraxis für Onkologie und Hämatologie, Köln, Germany 1

Introduction: WINHO developed a set of 46 quality indicators as a tool for self-monitoring, benchmarking and to enhance quality reporting in the context of outpatient care in medical oncology. Supported by German Cancer Aid, the indicators were generated in collaboration with oncology experts, incl. patient representatives (2009–2010), and evaluated regarding their feasibility (2010–2012). Subsequently, these quality indicators were tested in a pilot study (2012–2015). This presentation provides key results of the pilot study and critically evaluates the practicality of quality indicators in everyday practice. Methods: The pilot study consisted of 3 waves of data collection via an online questionnaire. Between 20 and 32 practices participated in each wave providing data from 58 patient records on average per practice per wave. The number of quality indicators examined in each wave varied between 6 (first wave) and 16 indicators (wave 2 and 3). Altogether, data were collected for 32 indicators from 4659 patient records in 37 oncology practices. Each practice received a feedback report with its results compared to anonymized results of other participating practices. Results: In general, the data indicate high levels of compliance concerning indicators of basic documentation compared to documentation of pain and psycho-social wellbeing. Stronger differences in adherence rates can be seen in indicators on therapy planning and implementation compared to basic documentation. However, adherence to quality indicators varies within as well as between practices. Due to the complexity of inclusion and exclusion criteria of some quality indicators (e.g. therapy planning), only small numbers of cases were obtained for these measures. In addition, collecting data for these indicators took more time than collecting data on basic documentation.

224

Oncol Res Treat 2015;38(suppl 5):1–270

Conclusions: Data collection and testing of the quality indicators were successful and prove the general applicability of WINHO indicators. The duration of data collection varies considerably between practices due to various documentation habits and IT systems. In order to be applicable in a broad manner, retrievability of data from patient records needs to be simplified. Facilitating improved documentation and IT systems in oncology practices would ease the collection and examination of the rich information documented by office-based cancer specialists. Disclosure: No conflict of interest disclosed. V751

Quality of life after cancer – how the extent of impairment is influenced by patient characteristics Peters E.1, Mendoza-Schulz L.2, Reuss-Borst M.1 Rehabilitation Clinic for Rheumatology and Oncology, Bad Kissingen, Germany, University Clinic for Psychiatry and Psychotherapie, Georg-August University, Göttingen, Germany 1 2

Introduction: Various factors influence lasting impairment of health related quality of life in oncological patients. Although this effect is well known, treatment methods have not yet been adapted broadly. The aim of this study was to identify patient characteristics which influence the impairment of quality of life and thus to identify those patients who need specific (psychosocial) interventions most urgently. Methods: 1879 cancer patients were filled in the EORTC QLQ C-30 questionnaire at the beginning of their inpatient rehabilitation. Patients’ scores were compared to those of 2081 healthy adults. Furthermore, differences in quality of life corresponding to sex, age, tumor site, TNM stage, interval between diagnosis and rehabilitation, and treatment method were examined. Results: Compared to the healthy population, the study group showed a decreased quality of life in all analyzed domains. This difference diminished with increasing age. Women reported a lower quality of life than men in general. Patients with prostate cancer showed the least impairment in several domains. Patients having undergone chemotherapy as well as radiotherapy were impaired the most. Surprisingly, TNM stage and interval between diagnosis and rehabilitation did not significantly influence quality of life. Conclusion: Despite an individualised and increasingly better tolerable therapy, the quality of life of cancer patients is still considerably impaired. Therefore, systematic screening of psychosocial aspects of cancer, e.g. quality of life, could identify those patients that need psychosocial interventions most urgently. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V752

Effective medical error prevention supported by an electronic central chemotherapy (CTx) management system Kaiser S.1, Reinhardt H.1, Szymaniak-Vits M.1, Otte P.1, Ruch M.2, Wolfrum P.1, Spadaro S.3, Opeker K.3, Wöhrl S.4, Rautenberg B.5, Stickeler E.5, Machein M.6, Brass V.7, Henke M.8, Meiß F.9, Hug M.4, Duyster J.1, Engelhardt M.3 Universitätsklinikum Freiburg, CCCF/Klinik für Innere Medizin I (Hämatologie und Onkologie), Freiburg, Germany, 2MPS – Medizinische Planungssysteme GmbH, Freiburg, Germany, 3Universitätsklinikum Freiburg, Klinik für Innere Medizin I (Hämatologie und Onkologie), Freiburg, Germany, 4 Universitätsklinikum Freiburg, Klinikumsapotheke, Freiburg, Germany, 5 Universitätsklinikum Freiburg, Klinik für Frauenheilkunde, Freiburg, Germany, 6 Universitätsklinikum Freiburg, Klinik für Neurochirurgie, Freiburg, Germany, 7 Universitätsklinikum Freiburg, Klinik für Innere Medizin II (Gastroenterologie, Hepatologie, Endokrinologie & Infektiologie, Freiburg, Germany, 8 Universitätsklinikum Freiburg, Klinik für Strahlenheilkunde, Freiburg, Germany, 9 Universitätsklinikum Freiburg, Klinik für Dermatologie und Venerologie, Freiburg, Germany 1

Introduction: Patients (pts) with cancer often receive complex medication regimens. This may lead to high medication error (ME) rates (Walsh, JCO 2008; Markert,Engelhardt, IJC 2009). Our central CTx management system (CMS) incorporates all important elements to avoid ME, namely a) detailed CTx protocols including co-medication, b) SOPs and clinical pathways, c) a multidisciplinary team and d) an electronic CTx ordering and monitoring system. The electronic CTx management tool (ChemoAS) has become an indispensable part of the clinical routine. We describe the reduction of ME and practical targets for intervention. Methods & Results: Chemo-AS is a web-based database application containing >600 standardized and regularly updated CTx protocols (in parts published in Das Blaue Buch; Engelhardt et al. 2014). Physicians generate CTx orders via Chemo-AS, which are processed by the clinical pharmacy (CP) and our Clinical Cancer Research Group (CCRG). A clinical accuracy check is performed by both CP and CCRG with different focus. Any detected error is instantly reported to the responsible physician and consequently corrected. If a CTx order is flawless, a detailed pt-individual treatment schedule created via Chemo-AS is sent to the ward and used as a valid prescription. Chemo-AS was first implemented at the department of Hematology&Oncology and has meanwhile been transferred to all other adult oncology units of our Comprehensive Cancer Center. In a prospective study of consecutive cancer pts, who received CTx between 2005 and 2014, all CTx orders/year were analysed (Table 1). Error rates considerably declined as a result of the implementation of Chemo-AS, our interdisciplinary team, continuous education and close collaboration with the CP. Due to our CMS, >99.9% of all errors were detected and eliminated, thus did not reach the pt. Tab. 1.

vides work simplification for physicians and nurses. A transfer to other interested cancer centers is currently in preparation with the project ´ChemoCompile´ via cooperation with an e-health company. Disclosure: No conflict of interest disclosed. V753

New scoring system allows prediction of outcome of ICU treatment in critically ill haematologic patients Reinbach M.-C.1, Kondakci M.1, Germing U.1, Kobbe G.1, Fenk R.1, Schroeder T.1, Rachlis E.1, Zeus T.2, Rassaf T.2, Haas R.1 Universitätsklinikum Düsseldorf, Klinik für Hämatologie, Onkologie und Klinische Immunologie, Düsseldorf, Germany, 2Universitätsklinikum Düsseldorf, Klinik für Kardiologie, Pneumologie und Angiologie, Düsseldorf, Germany 1

Introduction: Intensive care unit (ICU) support for critically ill patients suffering from a haematological malignancy is controversial because of the poor prognosis of this patient group. In this retrospective single centre study we evaluated the current status quo for ICU-, in-hospital-, 6-month- and 1-year mortality within our university hospital. Furthermore, we aimed at identifying factors predicting survival of critically ill haematological patients who were admitted to the ICU. Methods: Files of all patients treated at our haematology/ oncology department and admitted to one of the ICUs of the university hospital for at least 24 hours were retrospectively screened and relevant data were documented in a standardized manner. Collected data included patient characteristics, reason for ICU admission, cause of death, organ support therapy on ICU, and further haematological relevant values. Laboratory data were recorded within the first 24h of ICU admission. APACHE II score, SAPS II score and SOFA score were calculated using the collected data. Results: We analysed 228 (148 men, 80 women) patients treated at our university hospital during the 5-year period between 2009 and 2013. Median age was 57 years (range 14–89). ICU-, in-hospital-, 6-month-, and 1-year mortality was 51.3%, 62.3%, 72.3%, and 75.4%, respectively. APACHE II score, SAPS II score and SOFA score showed good predictive value regarding the ICU outcome, but are difficult to calculate in the daily clinical routine. We developed a new scoring system with the goal to separate survivors from non-survivors of the ICU- and in- hospital stay. The score is calculated on the basis of seven variables assigning one point for each variable, if values exceed or fall below following values: pH (≤7.21), lactate (≥7.0 mmol/l), potassium (≥5.7 mEq/l), bicarbonate (≤15.3 mEq/l), creatinine (≥3.0 mg/dl), bilirubin (≥3.0 mg/dl), and urin volume (≤1 l/24h). Patients with a score value of 0, 1–2, ≥3 showed ICU survival rates of 70.8% (80 of 113 patients), 31,7% (19 of 60) and 6,9% (2 of 29), respectively. Conclusion: In our retrospective analysis we developed a prognostic score for ICU outcome which is less complicated to calculate than the pre-existing other scores without any loss of power in separating survivors from non-survivors in our patient population. Its predictive power has to be confirmed in further prospective trials. Disclosure: No conflict of interest disclosed.

Conclusions: Our central CTx management system has shown to substantially improve the quality of CTx treatment and pt safety and it pro-

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

225


CONTENTS AUTHOR INDEX

Posterdiskussion

MPN / sonstige Hämatologie P754

Systematic evaluation of overall Quality of Life in patients with Philadelphia negative MPN: An analysis of the German SAL-MPN registry Kaifie A.1, Gattermann N.2, Wolf D.3, Hollburg W.4, Klausmann M.5, Maintz C.6, Hänel M.7, Gökkurt E.8, Göthert J.R.9, Platzbecker U.10, Parmentier S.11, Lang F.12, Hansen R.13, Isfort S.1, Serve H.12, Berdel W.E.14, Ehninger G.10, Brümmendorf T.H.1, Koschmieder S.1, Study Alliance Leukemia (SAL) Dept. for Hematology, Oncology, Hemostaseology and SCT, Faculty of Medicine, RWTH Aachen University, Aachen, Germany, 2Dept. for Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Duesseldorf, Germany, 3Internal Medicine 3, Oncology, Hematology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany, 4Practice for Hematology and Oncology Altona, Hamburg, Germany, 5Practice for Hematology, Oncology and Gastroenterology, Aschaffenburg, Germany, 6 Practice for Hematology and Oncology, Wuerselen, Germany, 7Dept. for Hematology, Oncology, Stem cell transplantation, Hospital Chemnitz, Chemnitz, Germany, 8Practice for Hematology-Oncology Eppendorf, Hamburg, Germany, 9 Dept. for Hematology, University Hospital Essen, Essen, Germany, 10Dept. for Hematology, University Hospital Dresden, Dresden, Germany, 11Dept. for Hematology, Oncology and Palliative Care, Rems-Murr-Hospitals, Winnenden, Germany, 12Dept. for Hematology and Oncology, University Hospital Frankfurt/ Main, Frankfurt/Main, Germany, 13Practice for Hematology and Oncology, Kaiserslautern, Germany, 14Dept. of Medicine A, University Hospital Muenster, Muenster, Germany

Conclusion: Though patients with different MPN subtypes suffer from similar symptoms, the overall impairment of QoL is significantly higher in PMF patients. Besides symptoms, typical complications of the underlying disease, in particular, bleeding and vascular events, significantly affect the patient’s QoL. Therapeutic and symptomatic strategies need to focus on both symptom relief and the reduction of complications in order to improve global QoL in MPN. Disclosure: Andrea Kaifie: No conflict of interest disclosed. Steffen Koschmieder: Advisory Role: Novartis, BMS, Pfizer, Ariad; Financing of Scientific Research: Novartis, BMS, Pfizer, Shire, Celgene, Ariad; Expert Testimony: Novartis, Joint Research Center Bayer and RWTH Aachen University, BMS, Novartis Foundation; Other Financial Relationships: Reisekostenübernahmen Novartis, BMS, Pfizer, Shire, Celgene, Ariad.

1

Introduction: Patients with Ph negative Myeloproliferative Neoplasms (MPN) often suffer from severe symptoms, resulting in an impairment of the quality of life (QoL). To assess QoL and its influencing factors in different MPN subtypes, we used an MPN specific questionnaire in our patient cohort. Methods: The SAL-MPN registry is a non-interventional prospective study. Inclusion criteria include: age of at least 18 years, diagnosis of an MPN according to WHO criteria (2008), and written informed consent. At baseline, all patients completed the standardized MPN-SAF-TSS questionnaire (ordinal scale, 0–10) with additional questions concerning further symptoms (vertigo, headache) and self-assessed global QoL. Clinical data concerning previous history of cancer, bleeding, and thrombosis/embolism were extracted from the registry. For statistical analysis, descriptive methods, Pearson correlation, and multiple linear regression analyses to predict global QoL were used. Results: Data of 275 patients (median age 65 years) were evaluated for this analysis. Among patients with MPN, PMF patients showed highest mean symptom severity for fatigue, vertigo, and concentration problems (Table 1). For fatigue, vertigo, abdominal discomfort, and early satiety statistically significant differences between the three MPN subtypes were detectable. Furthermore, the global QoL of PMF patients was markedly impaired in comparison to ET and PV (p < 0.05). Weight loss and fever only played a minor role in the symptom’s analyses. Intriguingly, multiple linear regression analyses for prediction of global QoL showed a significant influence for the symptoms fatigue, vertigo, abdominal discomfort as well as vascular and bleeding events in the multivariate model (R square = 0.55, p < 0.05).

P755

Evaluation of BRD4 as potential target in JAK2 V617F+ MPN cells Keller A.1, Peter B.1,2, Zuber J.3, Valent P.1,2, Hadzijusufovic E.1,2,4 Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria, 2Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria, 3Research Institute of Molecular Pathology, Vienna, Austria, 4Department of Companion Animals and Horses, Small Animal Clinic, Internal Medicine, University of Veterinary Medicine, Vienna, Austria 1

Myeloproliferative neoplasms (MPN), including polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF), are neoplasms defined by clonal expansion and accumulation of myeloid cells, erythrocytes and/or platelets in the bone marrow (BM) and other organs. In many patients, the JAK2 V617F mutation is found. Although the disease is chronic and indolent in most cases, fatal progression may occur. So far, the only curative approach for these patients is bone marrow transplantation. Therefore, current research is evaluating new drug targets. The epigenetic reader bromodomain-containing protein 4 (BRD4) has recently been identified as a promising target in acute myeloid leukemia. Therefore, we examined the potential value of BRD4 as a molecular target in MPN. We employed two JAK2 V617F+ cell lines, SET-2 and HEL, as well as BM samples obtained from MPN patients (ET: n = 4; PV: n = 2) in our experiments. Furthermore, three inhibitors of BRD4 were applied (JQ1, BI2536 and BI6727). As assessed by immunocytochemistry, SET-2 cells and HEL cells were found to express cytoplasmic BRD4. Expression of BRD4 mRNA in SET-2 and HEL cells was confirmed using qPCR. As assessed by 3H-thymidine uptake, all three BRD4 blockers were able to inhibit the proliferation of MPN cells with the following IC50 values (nM): SET-2: JQ1, 50–100; BI2536, 20–40; BI6727, 50–75; HEL: JQ1, 100–500; BI2536, 20–40; BI6727, 30–50; primary MPN cells: 500–1000 with all three drugs. Using flow cytometry, we examined the effects of the BRD4 inhibitors on induction of apoptosis and cell cycle arrests in SET2 and HEL cells. All tested inhibitors were able to induce apoptosis in a dose- and time-dependent manner, with ED50 values of >5 µM for JQ1 and 0.5–5.0 µM for BI2536 and BI6727. In cell cycle experiments, BI2536 and BI6727 were found to induce a G2/M phase arrest in both cell lines. JQ1 led to a G1 phase arrest in HEL cells, while showing no effect on cell cycle in SET-2 cells. To confirm the importance of BRD4 in MPN cells, knockdown experiments in SET-2 and HEL cells were performed using

Tab. 1. zu P754 Symptom severity and global QoL in PMF, PV, and ET

Symptom Severity, mean (SD)

Fatigue

Vertigo

Concentration

Abdominal discomfort

Bone pain

Early satiety

Weight loss

Fever

Global QoL

PMF n = 75

5.33 (3.29

3.53 (3.3)

3.4 (3.1)

2.88 (3)

2.56 (3.1)

2.46 (2.8)

1.3 (2.2)

0.11 (1.4)

4.21 (2.8)

PV n = 95

3.76 (2.8)

2.17 (2.6)

2.31 (2.7)

1.6 (2.4)

2.47 (3.1)

2.07 (2.9)

0.8 (2.2)

0.3 (1.4)

3.1 (2.2)

ET n = 80

4.46 (3.2)

2.73 (3.2)

2.49 (3.0)

2.05 (2.9)

2.74 (3.3)

1.28 (2.1)

0.58 (1.6)

0.29 (1.3)

3.3 (2.4)

p-value

0.0087

0.0278

0.0707

0.0221

0.8594

0.0238

0.1285

0.05642

0.0265

226

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

two different shRNAs against BRD4. In these experiments, both shRNAs were found to block cell proliferation. In summary, our data show that BRD4 is expressed in JAK2 V617F+ MPN cells and that targeting of BRD4 is associated with decreased proliferation and induction of apoptosis. The clinical implications of BRD4-suppression in MPN remain to be determined. Disclosure: No conflict of interest disclosed.

immunophenotype of HSPCs. This may reflect the physiological requirement to stimulate the remaining hematopoiesis. A better understanding of these soluble factors may provide better insight into the regulatory feedback process involved in MF. Disclosure: No conflict of interest disclosed. P757

P756

Serum of myelofibrosis patients enhances proliferation and maintenance of healthy hematopoietic stem and progenitor cells in vitro Lubberich R.K.1, Walenda T.1, Frobel J.1, Goecke T.W.2, Strathmann K.3, Koschmieder S.4, Wagner W.1

Interim results of the phase IV JAKoMo trial, a prospective, non-interventional study of symptoms and quality of life in myelofibrosis patients receiving ruxolitinib therapy Wehrle J.1, Geer T.2, Brudler O.3, Schäfer E.4, Tesch H.5, Cavanna D.6, Bachhuber P.6, Markhauser M.6, Koschmieder S.7, Mesa R.A.8, Pahl H.L.1 Uniklinik Freiburg / Innere Medizin / Hämatologie, Onkologie und Stammzelltransplantation, Sektion Molekulare Hämatologie, Freiburg, Germany, 2Medizinische Klinik III, Diakonie-Klinikum, Schwäbisch Hall, Germany, 3 Hämatologisch-onkologische Praxis Brudler/Heinrich/Bangerter, Augsburg, Germany, 4Onkologische Schwerpunktpraxis, Bielefeld, Germany, 5Onkologische Gemeinschaftspraxis am Bethanien-Krankenhaus, Frankfurt/Main, Germany, 6 Novartis Pharma GmbH, Nürnberg, Germany, 7Klinik für Hämatologie, Onkologie, Hämostaseologie und SZT, Med. Fakultät Uniklinik RWTH, Aachen, Germany, 8Mayo Clinic Cancer Center, Division of Hematology & Medical Oncology, Scottsdale, United States 1

Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany, 2Department of Obstetrics and Gynecology, RWTH Aachen University Medical School, Aachen, Germany, 3Institute for Transfusion Medicine, RWTH Aachen University Medical School, Aachen, Germany, 4Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany 1

Introduction: Myelofibrosis (MF) is a myeloproliferative neoplasm (MPN) associated with gradual replacement of normal bone marrow by fibrous tissue leading to progressive bone marrow failure. Several genetic aberrations in hematopoietic stem and progenitor cells (HSPCs) have been identified that contribute to MF. However, little is known about the underlying mechanisms that entail excessive fibrosis of the bone marrow and ineffective hematopoiesis. Furthermore, it is unclear whether or not soluble factors contribute to this process. Therefore, we investigated effects of MF serum on proliferation and maintenance of an undifferentiated phenotype of HSPCs in vitro. Methods: CD34+ HSPCs were isolated from human cord blood, labeled with carboxyfluorescein diacetate N-succinimidyl ester (CFSE) and subsequently cultivated in StemSpan medium supplemented with 10% of serum of either diseased or healthy donors (13 MF samples vs. 15 control samples). After five days of in vitro expansion, proliferation (CFSE) and immunophenotype (CD34, CD133, CD45) of HSPCs were analyzed by flow cytometry. The influence of MF Serum on colony forming unit potential is concurrently analyzed. Results: Addition of MF serum significantly enhanced proliferation of HSPCs in comparison to normal serum controls (p < 0.00005 in each of three biological replica). Subsequently, we analyzed mean fluorescence intensities within gates corresponding to specific numbers of cell divisions (CFSE). HSPCs cultured in MF serum had an overall increased expression of CD34 and this was particularly observed in slow dividing cells (p < 0.05). Furthermore cultivation with MF serum resulted in a higher expression of CD133 in the slow dividing fraction (p < 0.05), but converges to normal expression level in fast proliferating cells. CD45 expression was constantly higher in HSPCs cultured in MF serum in comparison to normal serum (p < 0.05). Conclusion: Serum from MF patients comprises soluble factors that significantly enhance proliferation and influence maintenance of a primitive

The oral JAK1/JAK2 inhibitor ruxolitinib was approved for the treatment of disease-related symptoms or splenomegaly in adult patients with primary myelofibrosis (PMF), post polycythemia vera MF (PPV-MF), or post essential thrombocythemia MF (PET-MF). Data detailing the effect of ruxolitinib on the quality of life of MF patients have not been obtained outside of clinical trials. The prospective, non-interventional phase IV study JAKoMo was therefore initiated to determine disease related symptoms and quality of life before and during ruxolitinib treatment in patients followed in the community setting. A cohort of 350 patients was monitored using the Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF). This validated self-assessment form allows patients to report symptom severity and quality of life in 19 areas. Patients scored the symptoms on a scale from 0 to 10, where 10 is the “worst imaginable” symptom severity. (Scherber R., Blood 2011) Based on these values, the MPN-SAF total symptom score (TSS) was calculated. (Emanuel RM, JCO 2012) Here, interim data is reported for those 225 patients for whom baseline data and at least one other time point were available. This sub-cohort did not differ in age, sex, or clinical parameters from the 350 patients comprising the entire JAKoMo cohort. Compared to baseline, patients reported a statistically highly significant decrease in TSS during ruxolitinib treatment at 1 month, which was maintained at 12 months (decrease of 9.67 and 10.2; n = 207 and n = 101, respectively; p < 0,0001 each). In particular, night sweats, pruritus and weight loss improved significantly, with decreases (± SEM) of 1.62 ± 0.29, 1.44 ± 0.30 and 2.74 ± 0.39 in the severity score at 12 months, respectively. The effect on symptoms not included in the TSS are depicted in Table 1. More detailed analysis including the correlation of clinical status with symptom self-assessment will be presented. Our data show that significant symptom improvement and an increase in overall quality of life can be achieved with ruxolitinib treatment in MF pa-

Fig. 1. zu P757 Effect of Ruxolitinib treatment at 1 and 12 months

Quality of life

Abdominal pain

Headache

Dizziness

Numbness

Insomnia

Sad mood

Sexuality problems

Cough

mean change at 1 month (SEM)

–1.12 (0.19)

–0.92 (0.19)

–0.34 (0.16)

–0.50 (0.18)

–0.39 (0.18)

–1.04 (0.21)

–0.80 (0.17)

0.04 (0.23)

–0.28 (0.16)

p (paired t-test: score at baseline vs 1 month)

<0.001

<0.001

0.0351

0.0078

0.0307

<0.001

<0.001

0.8523

0.0774

mean change at 12 months (SEM)

–2.74 (0.39)

–0.83 (3.15)

–0.44 (2.13)

–0.27 (0.28)

–0.04 (0.33)

–1.04 (0.37)

–0.53 (0.26)

–0.09 (0.37)

–0.04 (0.37)

p (paired t-test: score at baseline vs 12 month)

<0.001

0.0102

0.0413

0.3438

0.9015

0.0057

0.0488

0.8092

0.8824

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

227


CONTENTS AUTHOR INDEX

tients in daily clinical practice in Germany. Moreover, in the large majority of patients, this improvement is seen within the first month of treatment. Disclosure: Julius Wehrle: Advisory Role: Novartis. Heike Pahl: Advisory Role: Novartis; Financing of Scientific Research: Novartis. P758

Zeb2 loss in adult mice results in a myeloproliferative disorder and is associated with altered JAK/STAT signaling pathway Li J.1, Riedt T.1, Goossens S.2,3, Gütgemann I.1, Huylebroeck D.4, Haigh J.2,3, Brossart P.1, Janzen V.1 University Hospital Bonn (UKB), Bonn, Germany, 2Ghent University, Ghent, Belgium, 3Monash University, Melbourne, Australia, 4Katholieke Universiteit, Leuven, Belgium 1

Introduction: Zeb2, a member of the zinc-finger E-box-binding (ZEB) family transcription factors, has predominantly been characterized as a transcriptional repressor and plays an important role during embryonic development as an epithelial to mesenchymal transition (EMT) modulator and was also associated with tumor progression and metastasis. Our previous work showed that deletion of Zeb2 within hematopoietic compartment resulted in early embryonic lethality due to differentiation and migration defects. However, very little is known about the physiological role of Zeb2 in adult hematopoiesis. Methods: Using the interferon sensitive Mx1-Cre based inducible knockout model we investigated the composition of hematopoietic cells in blood, bone marrow and spleen upon Zeb2 deletion. In addition we analyzed signaling activity of JAK/STAT and MAPK signaling pathways under physiological conditions in-vivo and under stimulation with cytokines ex-vivo, as well as its inhibition by Ruxolitinib. Results: Upon induction of Zeb2 deficiency within the hematopoietic system we observed reduced erythrocytes and thrombocytes while granulocytes were increasing over time. In addition these mice developed a significant and progressive increase in spleen size. Histological analyses revealed a massive destruction of spleen architecture and a massive extramedulary hematopoiesis in Zeb2 depleted animals. Silver staining of bone marrow sections showed a significant increase in the appearance of reticular fibers in Zeb2Δ/Δ Mx1-Cre mice. In addition, there was an increase in frequency of megakaryocytes with abnormal morphology in the bone marrow and spleen. All above features have been described as a hallmark of myeloproliferative diseases in humans. Western blot analyses revealed a higher phosphorylation of Stat3 and Stat5 in Zeb2-deficient bone marrow cells, which was dramatically augmented upon cytokine stimulation, which was completely abrogated by Ruxolitinib. In-vivo treatment with Ruxolitinib by oral gavage for two weeks could significantly reduce spleen size in Zeb2 KO mice, but had little effect on controls. Conclusions: Zeb2 ablation in the hematopoietic compartment led to a phenotype with several features reminiscent of myeloproliferative disorders in human, such as bone marrow fibrosis, splenomegaly and extramedullary hematopoiesis. These phenotypes may in part be attributed to altered JAK/STAT signaling activity. Disclosure: No conflict of interest disclosed.

P759

Results of a phase I, single dose, randomized, 3-way crossover study, to assess the bioavailability of a novel anagrelide extended release (ER) formulation in com-parison to a commerically available anagrelide reference product (CARP) in healthy volunteers Petrides P.1, Zagrijtschuk O.2, Klade C.2 Hematology Oncology Center Munich & Ludwig Maximilians University Medical School Munich, München, Germany, 2AOP Orphan Pharmaceuticals AG, Wien, Austria 1

It has been observed that commercially available anagrelide formulations differ in their tolerability, especially with regard to frequency and intensity of vascular and cardiac adverse events. This difference is potentially related to the pharmacokinetics of the formulations: lower Cmax and AUC were associated in one formulation with better tolerability without compromising the platelet reduction effect [1]. This observation has led to the development of an ER formulation of anagrelide, searching for a better tolerability and extended therapeutic effect, with the potential consequence of fewer treatment drop-outs and improvement of the long-term patient compliance and treatment outcomes. Randomized, single dose, 3-way (fasting and fed conditions vs. reference) crossover study in 30 healthy volunteers was carried out. Plasma concentration of anagrelide was determined using validated methods, and the classical pharmacokinetic parameters were calculated. Usual safety data were collected and analyzed. It has been shown that the bioavailability of anagrelide ER is lower with regard to both rate and extent of absorption when compared to CARP. Absorption of the ER formulation was enhanced considerably when administered with a high-fat meal (data not shown), resulting in a biphasic absorption pattern. The AUC0-∞ expressed as µg/h/L (mean and min/max, resp.) was 16.39 (6.1/37.0) and 28.35 (11.9/74.4), Cmax in µg/L (mean and min/max, resp.) was 2.43 (0.8/8.0) and 9.89 (5.1/22.9), tmax (in h, median and min/max, resp.) was 8.00 (4.0/12.0) vs. 1.18 (0.7/2.5), the t1/2 (in h, median, min/max) was 3.88 (1.6/24.5) compared to 1.52 (0.9/7.3) for the ER formulation and CARP, resp.Tolerability was improved considerably when 2 mg anagrelide was taken as the ER form: a total of 13 adverse events (AEs) vs. 27 AEs with the CARP was observed. Direct time-relationship between the onset of AEs and maximum plasma concentrations (Cmax) was also observed. Observed improved safety of the ER formulation when compared to the licensed CARP formulation supports the validity of the relationship between PK profile and tolerability. Importantly, the improved tolerability was already apparent in this small, single-dose setting; the effect is expected to be more pronounced in the potential long time treatment of patients. Reference: 1 Petrides PE et al.: Clin Ther.31;2009:386–398. Disclosure: Petro Petrides: Expert Testimony: AOP Orphan Pharmaceuticals AG. Christoph Klade: Employment or Leadership Position: AOP Orphan Pharmaceuticals AG. P760

The JAK-inhibitor ruxolitinib exerts substantial impact on body weight Vonnahme M.1, Trebicka J.2, Isfort S.3, Koschmieder S.3, Brümmendorf T.H.3, Brossart P.1, Wolf D.1 University Clinic Bonn (UKB), Department of Internal Medicine III, Oncology, Hematology and Rheumatology, Bonn, Germany, 2University Clinic Bonn (UKB), Department of Internal Medicine I, Gastroenterology and Endocrinology, Bonn, Germany, 3RWTH Aachen University Hospital, Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, Aachen, Germany 1

Background: The JAK1/2 inhibitor ruxolitinib has substantial impact on symptom burden in myeloproliferative neoplasms (MPN) and patients significantly gain weight during therapy, which was related to reversal of

228

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

the katabolic state of myelofibrosis (MF) patients and normalization of the deregulated metabolic/adipokine milieu. In this retrospective analysis, we analysed the body weight (BW) and body mass Index (BMI) course during MPN history and the time before diagnosis. Patients and methods: We analysed BW and BMI from 45 patients with primary or secondary MF at defined time points during their disease history, in order to evaluate the impact of ruxolitinib on the body composition. From these 45 patients, 29 were treated with ruxolitinib due to symptomatic MF. Results: The mean BW loss from prior to diagnosis to treatment initiation was 3.38 kg in all patients, whereas BW loss was more pronounced in patients receiving ruxolitinib (minus 5.06 kg). This translates into a BMI change from 25.54 to 24.61 in the overall cohort and from 26.84 to 25.36 in ruxolitinib-treatetd MF. BW increased during ruxolitinib therapy (mean BW gain of 8.14 kg from initiation of therapy). This increase also translates into a rising BMI from 25.36 to 28.22. We noticed that individual patients showed a massive BW gain and thus compared the maximum BW during ruxolitinib treatment to their BW before MPN diagnosis. Strikingly, there was a significantly higher maximum BW during ruxolitinib treatment as compared to the individual BW of the patient prior to MPN diagnosis (75.19 kg vs. 83.21 kg; p = 0.01). Accordingly, the BMI was 25.54 prior to MPN diagnosis compared to 28.22 during ruxolitinib (p = 0.011). We also analyzed various metabolic parameters (cholesterol, HDL cholesterol, LDL cholesterol and HbA1c). Interestingly, despite that fact that patients gained BW above their BW prior to MPN diagnosis, we could not detect any negative impact on the lipid status of ruxolitinib-exposed patients. Conclusion: Given the limitation of the small sample size and retrospective nature of this analysis, BW gain may be relevant side effect during ruxolitinib therapy. This may be of particular importance in patients not losing too much BW during the course of their MPN (such as PV patients), because increased BW is an established risk factor for many cardiovascular diseases and cancer.

afflicted with more comorbidities (median CIRS score: 13 (p = 0.0206) compared to other pts (Median age: 62 yrs; median CIRS score: 5). Only 1/31 pts (3.2%) with MF would have been eligible for the COMFORT 1 trial, only 2/31 pts (6.5%) for the COMFORT 2 trial. Eligibility of PV pts for the RESPONSE trial was 2/25 pts (8%). Reasons for eligibility failure are listed in Table 1. Conclusions: On average, only 6% of our pts were eligible for the clinical trials analyzed. Although some criteria may have allowed individual case discussions, the majority of “real life” pts was not represented. In addition to recruitment aspects this analysis points out the necessity of further investigations to assess tolerability and safety of these drugs outside of controlled clinical trials. Tab. 1. Reasons for patients non-eligibility

P761

Isfort S.T.1, Kaifie A.1, Jost E.1, Schmitt K.1, Brümmendorf T.H.1, Koschmieder S.1 Uniklinik Aachen, Klinik für Hämatologie, Onkologie, Hämostaseologie und Stammzelltransplantation, Aachen, Germany 1

Introduction: Approval of the JAK1/2 inhibitor ruxolitinib has broadened treatment options for MPN patients (pts), but it was mainly based on a highly selected patient cohort (201 pts with myelofibrosis, 110 pts with PV + 150 pts who underwent cross-over). In order to evaluate congruency of the “real life” and clinical trial cohorts, we performed an analysis of study eligibility of our own patient cohort based on the criteria of the three MPN trials (COMFORT 1 and 2, RESPONSE). Methods: 56 MPN pts with polycythemia vera (PV; n = 25) and myelofibrosis (MF n = 31; n = 19 primary MF, n = 9 Post-PV-MF, n = 3 with Post-Essential Thrombocythemia (ET) – MF) presenting to our department during the past five years were evaluated according to the three clinical study protocols (NCT01243944, NCT00952289, NCT00934544). Inclusion/exclusion criteria were assigned to different categories (prior therapy/therapy options, risk profile/symptom quality, non-hematologic laboratory abnormalities [serum chemistry], hematologic laboratory abnormalities, patients´ performance status, concomitant disease/ concomitant medication). The Cumulative Illness Rating Scale (CIRS) was used for quantification of pts´ comorbidities. Results: Median age of pts at time of therapy decision was 62.5 yrs. PostET-MF group was older (median age: 72 yrs (P = 0.1594) and significantly

Abstracts

COMFORT 1 trial NCT00952289 N = 31 Median CIRS 5

COMFORT 2 trial NCT00934544 N = 31 Median CIRS 5

Prior therapy/therapy options

21 (84%)

24 (77%)

13 (42%)

Risk profile/ symptom distinction

19 (76%)

22 (71%)

15 (48%)

Non-hematologic laboratory abnormalities

4 (16%)

9 (29%)

9 (29%)

Hematologic laboratory abnormalities

NA

6 (19%)

6 (19%)

Patients´ performance status

1 (4%)

0

0

Concomitant disease/ concomitant medication

2 (8%)

11 (35%)

11 (35%)

Disclosure: No conflict of interest disclosed.

A high fraction of a “real world” cohort of patients with myeloproliferative neoplasms (MPN) would have failed enrollment into JAK1/2 inhibitor phase 3 clinical trials (More expanded access programs are needed to evaluate safety and efficacy of those medications in a real life setting)

RESPONSE trial NCT01243944 N = 25 Median CIRS 6

Disclosure: Susanne Isfort: Financing of Scientific Research: Pfizer, BMS; Other Financial Relationships: Reisekostenfinanzierung Roche, Mundipharma, Hexal, Amgen. Steffen Koschmieder: Advisory Role: Novartis, BMS, Pfizer, Ariad; Financing of Scientific Research: Novartis, BMS, Pfizer, Shire, Celgene, Ariad; Expert Testimony: Novartis, Joint Research Center Bayer and RWTH Aachen University, BMS, Novartis Foundation; Other Financial Relationships: Reisekostenübernahmen Novartis, BMS, Pfizer, Shire, Celgene, Ariad. P762

Uncommon reason of eosinophilia in peripheral blood smear Austein T.1, Schulz K.1, Back W.2, Kerstan H.3 St. Bernhard Hospital, Medizinische Klinik II-Abteilung für Hämatologie/intern. Onkologie, Brake, Germany, 2Pathologisches Institut, Bremerhaven, Germany, 3 St. Bernhard Hospital, Medizinische Klinik I-Abteilung für Allgemeine Innere Medizin, Brake, Germany 1

Introduction: Eosinophilia in peripheral blood smear isn`t an own disease, but rather a symptom. The differential diagnosis of eosinophilia are manifold. Mostly there were an underlying allergic or parasitic affection (Table 1). • Allergic (asthma bronchiale, rhinitis allergica, urticaria a.o.) • Parasitic infestation (worms, amebae, trichinae a.o.) • Dermatitis • Virus infection (CMV, EBV a.o.) • Gastrointestinal disorder (M. Crohn, colitis ulcerosa a.o.)

Oncol Res Treat 2015;38(suppl 5):1–270

229


CONTENTS AUTHOR INDEX

• Myeloproliferative neoplasm (CML, polyzythaemia vera, hypereosinophilic Syndrom a.o.) • Connective tissue diseases (Churgh-Strauss-syndrom, lupus erytematodes a.o.) • Malignant complaint (M. Hodgkin, AML M4Eo, cancer a.o) Tab. 1.

allergic diseases

e.g. bronchial asthma, urticaria

parasites or fungal infestation

e.g. worms, amebae, trichinae

dermatitis

virus infection

e.g. CMV, EBV

gastrointestinal disorders

e.g. Crohn´s disease, ulcerative colitis

myeloproliferative diseases

e.g. chronic myelogenous leukaemia, polyzythämia vera, hypereosinophilic syndrome

connective tissue diseases

e.g. systemic lupus erythematodes, Churgh-Strauss-syndrome

malignant neoplasia

e.g. Hodgkin´s disease, AML M4Eo, cancer

Differential diagnosis of eosinophilia Manual Hämatologie 2013 R. Fuchs et. al. Nora- Verlag Here we discribe a rare and uncommon reason of a peripheral eosinophilia. Case report: An 80.-year old male was admitted in the emergency room because of deteriation of his general condition. The blood count showed a mild leucocytosis and anaemia as soon as a severe thrombocytopenia (8.000/µl). The peripheral blood smear revealed 40% of atypical eosinophil granulocytes (Fig. 1.).

Fig. 2. Bone marrow smaer (CD 117 Staining).

Biopsies of stomach, bowel and colon were without mastcell infiltrates. The tryptase level was with 445 µg/l (normal < 11,5 µg/l) increased. Genetic analysis for c-KIT (816V), JAK-2 mutation, bcr-abl and FIP1L1-PDGFR Alpha were negative. We started first a therapy with 100mg prednisolon daily followed by 400mg imatinib per day, but only one month later the patient died in refractory disease. Conclusions: In case of eosinophilia also a systemic mastocytosis should be considered. Therefore immediately a bone marrow biopsy and tryptase analysis of the serum fluid are needed. Disclosure: No conflict of interest disclosed. P763

CBL is a critical negative regulator of megakaryopoesis Saur S.J.1, Märklin M.1, Ganser M.1, Poljak A.1, Kanz L.1, Kopp H.-G.1, Müller M.R.1 Universitätsklinik Tübingen, Abteilung für Hämatologie und Onkologie, Tübingen, Germany 1

Fig. 1. Peripheral blood smear (Pappenheim staining 1000×).

Examination appropiate disease in table 1 was not effective. Only then the bone marrow investigation resulted in unexspected systemic mastocytosis (Fig. 2.).

230

Oncol Res Treat 2015;38(suppl 5):1–270

Megakaryopoiesis is controlled by a variety of hematopoietic growth factors in order to maintain a physiological level of circulating platelets. Thrombopoietin (TPO) is the main regulator of megakaryopoiesis modulating megakaryocyte differentiation, promoting endomitosis and proplatelet formation. To allow proper proliferation and differentiation of different hematopoetic lineages, TPO signal transduction must be tightly regulated. Several mechanisms negatively modulate hematopoiesis and differentiation of the megakaryocytic lineage like cytokine signaling or protein phosphatases. However, one of the most effective mechanisms to permanently disable activated signaling proteins is by targeted degradation via lysosomes or proteasomes. In this study, we investigated the mechanisms that regulate TPO-mediated degradation of the thrombopoietin receptor (c-Mpl) in primary mouse cells. Previous studies have identified CBL as an E3 ligase responsible for the ubiquitination of c-Mpl in cell lines. In order to determine the potential role of CBL in murine thrombopoiesis, we used a conditional PF4-Cre CBL knockout mouse model to specifically delete CBL in the megakaryocytic lineage. Mice without a CBL deletion served as controls. The experimental cohort showed significantly higher numbers of megakaryocytes in the bone marrow and of platelets in the peripheral blood as compared to control mice. In addition, the platelets from the CBL ko mouse strain were of significantly smaller size (43 vs. 38 fL, p = 0.0022). To evaluate the role of CBL in mature megakaryocytes, total bone marrow was grown in TPO-containing culture medium for 72 hours. Mature megakaryocytes were eventually isolated on a BSA-density gradient. A subse-

Abstracts


CONTENTS AUTHOR INDEX

quently performed Western Blot analysis revealed a significant reduction of c-Mpl ubiquitination in the CBL ko mice as compared to control mice, thereby identifying CBL as a critical negative regulator of megakaryopoesis. Taken together, we have successfully ablated CBL specifically from the megakaryocyte lineage and could demonstrate that this has profound effects on platelet counts and size. In addition, we showed that CBL ablation leads to reduced ubiquitination of c-Mpl and a consecutively longer half life of this protein culminating in substantially increased megakaryopoiesis in the CBL ko kohort. In summary, these data enhance our understanding of the regulation of TPO signaling and the physiological role of CBL in the megakrayocytic lineage. Disclosure: No conflict of interest disclosed. P764

Interlaboratory comparison (“round robin“) of diagnostic quality in haematological cytomorphology (INSTANDDüsseldorf) Rotermund M.1, Gassmann W.1 St.Marienkrankenhaus Siegen, Hämato-/Onkologie, Siegen, Germany

1

Introduction: The cytomorphology of haematological diseases is an important diagnostic tool. The focus of this interlaboratory comparison (“round robin” test) is diagnostic quality. 1772 single bone marrow specimens were prepared and evaluated. These specimens encompass the performance level of experienced participants. Results were categorized according to the nature of the mistake and the severity of the disease. Methods: The selection and evaluation of the specimens was initally conducted by a cytologist from a reference laboratory. The participants were to submit a cytomorphological diagnosis with the help of a questionnaire and little additional information. In conclusion, the participants received a detailed error analysis and explanatory video prints of the specimens. This analysis was restricted to laboratories achieving more than 70% of the total score. Results: The error rates were spread over a severity scale of 4 levels (range between 0.0 to 35,1%). Error rates were unusally high for non-pathological specimens (10,3%) and myeloproliferative neoplasias (10,0%), the latter despite straightforward clinical information such as a thrombocytosis over an extended period of time. The percentage of false diagnoses in hemolytic anemia were 13,9%, in megaloblastic anemia 10,3% within which most cases were confounded with myelodysplasia. Myelodysplastic cases showed error rates up to 23,5%. MDS-subtypes error rates were RARS (11,5%), RAEB (35,1%) and 5q-minus-syndrome (34,6%). In acute myeloid leukemia errors appeared in 5,6%. The Remission status in acute myeloid leukemia (7,7%) and acute lymphoblastic leukemia(10,6%) is a challenge, especially in AML M4Eo. In this particular case only 18,5% of the participants diagnosed an early relapse. From a different point of view when participants finally diagnosed a case, such as MDS type RAEB, results were correct in 92% of cases. In contrast, in the case of reactive changes the result was correct in only 19.5%. Conclusion: In addition to offering extensive training and continued education programs, a panel of experts, such as in the ongoing CLL-/AMLstudy group trials, should be available to determine reference diagnoses. Modern digital media can simplify the necessary work routines.

P765

Valproic acid enhances the immunosuppressive capacity of human mesenchymal stromal cells Killer M.C.1, Brendel C.1, Neubauer A.1, Nold P.1 Philipps University of Marburg, Department of Hematology, Oncology and Immunology, Marburg, Germany 1

Introduction: Human mesenchymal stromal cells (MSC) are currently under intensive investigations due to their immunomodulatory capacities. Amongst others, they are reported to distinctly inhibit the proliferation of human Tcells. Therefore, they are a promising tool in stem cell therapy, e. g. for the treatment of autoimmune diseases or Graft-versus-host disease (GvHD) after allogeneic cell transplantations. Methods: We investigated human MSC regarding their general ability for immunomodulation via a coculture assay with human peripheral blood mononuclear cells (PBMC). The proliferative capacities of stimulated Tcells, with and without addition of MSC, were measured via flow cytometry. The histone deacetylase (HDAC) inhibitor valproic acid (VPA) was added to the coculture in order to analyze possible effects on the interaction between MSC and Tcells. Results: In our hands, MSC were generally able to suppress Tcell proliferation, but the outcome of the coculture experiments depended on both the MSC and the PBMC donor. Addition of VPA as well as pretreatment of MSC with VPA enhanced MSC-mediated Tcell suppression. Moreover, a potent direct inhibitory effect of VPA on Tcells was observed. Conclusion: VPA is a widely prescribed and well-tolerated drug used for the treatment of e. g. seizure disorders, migraine and bipolar disorders. As we showed its ability to enhance the immunomodulatory capacities of human MSC, it could be a promising tool for MSC priming in order to enhance their capabilities as an immunosuppressive cellular product. In addition a concomitant application of VPA together with MSC appears as a conceivable strategy for improvement of cellular therapy for GvHD.

Disclosure: No conflict of interest disclosed.

Fig. 1. Effects of VPA on T-cell proliferation

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

231


CONTENTS AUTHOR INDEX

P767

Results of the non interventional Observational study OncoBOS in chemotherapy-induced anaemia in ONCOHaematological patients treated with Binocrit® – OncoBOSstudy – an Austrian multi-center experience Vogl U.M.1, Schreieck S.2, Ludescher C.3, Pecherstorfer M.4, Öhler L.1, Haslbauer F.5 St. Josef Krankenhaus Wien, Interne I, Onkologie, Wien, Austria, 2BKH Reutte, Innere Medizin, Reutte, Austria, 3Ambulatorium Hämatologie/Onkologie Innsbruck, Innsbruck, Austria, 4LKH Krems, Innere Medizin, Krems, Austria, 5 Krankenhaus Vöcklabruck, Innere Medizin, Vöcklabruck, Austria 1

Fig. 2. Disclosure: No conflict of interest disclosed. P766

Remission of refractory Rosai-Dorfman disease after chemotherapy with clofarabine Kreißelmeier K.-P.1, Hinterleitner C.1, Müller K.2, Heissner K.1, Kanz L.1, Kopp H.-G.1, Müller M.R.1 Universitätsklinik Tübingen, Innere Medizin II – Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2Universitätsklinik Tübingen, Innere Medizin III – Kardiologie und Kreislauferkrankungen, Tübingen, Germany 1

Background: Rosai-Dorfman disease (RDD, sinus histiocytosis with massive lymphadenopathy) is a rare idiopathic nonmalignant histiocytic disorder. It typically occurs in children, adolescents or young adults, presenting with massive cervical lymphadenopathy. RDD can show a variable clinical course with repeated exacerbations and spontaneous regressions. However, fatal outcomes can occur, and extranodal involvement has been reported to predict a poor outcome. While treatment with steroids has been used as first-line therapy, in refractory cases a variety of chemotherapeutic agents have been used with frequent disappointing results. Clinical case: A 25-year-old man presented with enlargement of cervical lymph nodes, arthralgia, repeating episodes of fever and cutaneous nodules. Multiple biopsies of bone marrow, skin and lymph nodes remained non-diagnostic and symptomatic treatment led to no significant clinical improvement. After a biopsy of enlarged nasal tissue, a diagnosis of Rosai-Dorfman disease with cervical lymphadenopathy and extranodal involvement of the skin and nose was made. Steroid therapy resulted in only minor regression of lymphadenopathy with early relapse, accompanied by fever, massive weight loss and additional involvement of multiple bone sites and lacrimal glands. Treatment with four doses of rituximab brought no improvement. While undergoing treatment with imatinib which has been described to be efficient in a subset of patients with RDD, lymphadenopathy progressed and involvement of kidneys and bone marrow occurred with further clinical deterioration. Subsequently, a salvage therapy with clofarabine was started. After two courses of clofarabine substantial regression of nodal and extranodal disease could be achieved. The patient´s overall condition improved significantly and fever subsided. A total of six cycles of clofarabine were applied with progressive improvement of clinical presentation and normalization of body weight. A PET/ CT scan showed complete regression of extranodal sites and a nearly complete regression of nodal involvement with minimal remaining activity. The treatment was well tolerated but resulted in prolonged pancytopenia with repeated need for blood and platelet transfusions and multiple hospital admissions for febrile neutropenia. Conclusions: Clofarabine is an effective salvage therapy for the treatment of refractory Rosai-Dorfman disease. Treatment was well tolerated but can result in prolonged pancytopenia. Disclosure: No conflict of interest disclosed.

232

Oncol Res Treat 2015;38(suppl 5):1–270

Introduction: Chemotherapy associated anaemia is a common event during cancer treatment. Anaemia has a considerable impact on the quality of life of cancer patients. Around 75% of patients develop anaemia during chemotherapy treatment and therefore need either blood transfusions or erythropoietins (EPO). Binocrit® is an Epoietin-alpha biosimilar approved for the treatment of cancer associated anaemia and for anaemia secondary to chronic renal failure. Methods: Patients with solid tumors or lymphomas over the age of 18 presenting with chemotherapy associated anaemia deemed necessary to receive Binocrit® by the treating physician, were included in the study. Patients’ demographics and laboratory results were collected at baseline, after four and twelve weeks of treatment with Binocrit®. The main key factors for treating chemotherapy-associated anaemia and for prescribing Binocrit® were collected. Results: We present the data of 63 patients (male: n = 29, female: n = 34) with a median age of 68 years from 5 Austrian centers who were treated with Binocrit® for chemotherapy-associated anaemia as determined by the treating physician. More than half of the patients were in good performance status (Karnofsky Performance Score >80%). The most common tumor site was breast, followed by lung and pancreatic cancer. Over 98% received palliative chemotherapy, mainly in a first-line setting. 16% of patients received blood transfusions before the start of Binocrit® and 22% during Binocrit® treatment. The median start dose of Binocrit® was 30 000 IU per week. The median haemoglobin level at start of Binocrit® was 8.8 g/dL. We report a significant improvement of haemoglobin levels during Binocrit® treatment to 10.75 g/dL at week 12 (p < 0.05). Three SAEs were reported. Of those two patients died during the follow-up period due to tumor progression. The SAEs were not deemed related to Binocrit®. The physicians’ main key factors for treating anaemia were combating fatigue, maintaining quality of life and reducing the number of blood transfusions. The main reasons for prescribing the EPO Binocrit® were product efficacy, improved control of increase of haemoglobin and cost reduction. Conclusion: Binocrit® improves haemoglobin levels significantly after twelve weeks of treatment and has an excellent safety profile and good cost efficiency. Disclosure: Ursula Vogl: Advisory Role: BAyer; Financing of Scientific Research: Roche, Janssen-Cielag, Bayer, Novartis, Pfizer, Sandoz. Ferdinand Haslbauer: No conflict of interest disclosed. P768

Defective primary hemostasis as a cause of bleeding in patients with plasma cell disorders Hinterleitner C.1, Kreisselmeier K.-P.1, Pecher A.-C.1, Weisel K.1, Kanz L.1, Kopp H.-G.1, Jaschonek K.G.1 Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, Eberhard-Karls University, Tübingen, Germany 1

Introduction: Paraproteinemia has been associated with both, thromboembolic and bleeding complications. In previous retrospective analyses a direct correlation of serum immunoglobulin levels (especially IgM and IgA) with critical bleeding events was shown. Underlying mechanisms include interference with platelet function, immune thrombocytopenia, clotting factor deficiencies and acquired von Willebrand syndrome (AVWS).

Abstracts


CONTENTS AUTHOR INDEX

Methods: Laboratory data, including qualitative and quantitative analyses of von Willebrand Factor, collagen binding assay (CBA) and PFA-100 closure time from 97 patients with plasma cell disorders between 2003 and 2015 were retrieved and cases were evaluated retrospectively for bleeding complications. Coagulation abnormalities were correlated with clinical history and disease parameters. Results: 50 out of 97 patients (51%) displayed bleeding complications including multiple hematomas (n = 26), epistaxis (n = 8), or perioperative bleeding (n = 6). Bleeding vs. non-bleeding patients showed no differences regarding age, gender, previous chemotherapy, or immunoglobulin isotypes. Special coagulation lab results of patients with bleeding events are shown in table 1. Elevated serum free light-chain, but not complete Ig paraprotein levels were significantly associated with bleeding events (p < 0.001), prolonged PFA-100 closure time (p < 0.001), and pathological CBA/vWF-ratio (p < 0.001). Conclusion: Bleeding events were most commonly due to defects of primary hemostasis, i.e. global coagulation tests were insufficient in predicting bleeding. In contrast, when taken PFA-100 and CBA/vWF-ratio together, 68% of the bleeding patients could be detected. As vWF:Ag and RCo are unreliable in determining AVWS we suggest that both, PFA100 and CBA/vWF-ratio should be considered in patients with plasma cell disorders and signs of bleeding. Furthermore, serum free light-chain levels were correlated with hemorrhage and detectable AVWS. Therefore, free Ig light-chains might play a role in the pathophysiology of bleeding.

color flow cytometry and molecular analysis of MYD88 (L265P) mutation of bone marrow aspirates or peripheral blood were performed. Results: In five patients, we identified a B-cell clone (CD19+, k/l restricted); four of these patients showed a MYD88 (L265P) mutation. In three patients a clonal plasma cell population (CD38+, CD138+, SLAMF7+, k/l restricted) was found, among them one patient was positive for the MYD88 (L265P) mutation. In two patients a clonal population with both B-cell and plasma cell immunophenotype (CD19+, CD38+, SLAMF7+, CD138+, k/l restricted) was identified, both were negative for MYD88 (L265P) mutation. Conclusion: In patients with IgM monoclonal gammopathy a B-cell, plasma cell or a mixed phenotype clonal population could be identified by multicolor flow cytometry as the underlying cell clone. The MYD88 (L265P) mutation was not therefore strictly associated with a certain immunophenotype.

Tab. 1. Pathological coag labs in 50 patients wit

1

Normal

Abnormal

Patients (n)

Sensitivity (95% CI)

Specificity (95% CI)

PPV (95% CI)

NPV (95% CI)

PT, aPTT

50

0

50

0

0.94 (0.8–0.98)

0

0.47 (0.4–0.6)

vWF: Ag or RCoF

44

6

50

0.12 (0–0.3)

1 (0.9–1)

1 (0.5–1)

0.52 (0.4–0.6)

PFA-100

21

29

50

0.58 (0.4–0.7)

0.94 (0.8–0.98)

0.9 (0.7–1)

0.68 (0.6–0.8)

CBA/ vWF

34

16

50

0.32 (0.2–0.5)

0.98 (0.9–1)

0.94 (0.7–1)

0.57 (0.3–0.7)

PFA-100 or CBA/ vWF

16

34

50

0.68 (0.5–0.8)

0.94 (0.8–1)

0.92 (0.8–1)

0.73 (0.6–0.8)

Disclosure: No conflict of interest disclosed.

Posterdiskussion Multiples Myelom I P769

Characterization of clonal lymphocytes in patients with IgM monoclonal gammopathy Lisenko K.1, Hillengass J.1, Maier B.1, Schönland S.1, Hegenbart U.1, Ho A.D.1, Goldschmidt H.1, Hundemer M.1 Universitätsklinik Heidelberg, Hämatologie, Onkologie, Rheumatologie, Heidelberg, Germany 1

Background: Among patients with monoclonal gammopathy the immunoglobulin isotype IgM can be identified in 15–20%. Either clonal B-cells or plasma cells might represent the source of monoclonal IgM. Flow cytometry and molecular analysis of MYD88 might facilitate the identification of the underlying source of monoclonal IgM, i.e. B-cell or plasma cell origin. Methods: Ten patients with IgM monoclonal gammopathy associated with waldenstrom´s macroglobulinemia (n = 4), extranodal marginal zone lymphoma (n = 1), systemic AL-amyloidosis (n = 2), smoldering myeloma (n = 1) and monoclonal gammopathy of undetermined significance (n = 2) were included. To characterize the lymphatic clone, multi-

Abstracts

Disclosure: Katharina Lisenko: No conflict of interest disclosed. Michael Hundemer: Expert Testimony: Celgene. P770

NF-κB activity in bone marrow stromal cells is upregulated by lactate exchange with multiple myeloma cells Berenstein R.1, Wächte M.1, Nogai A.1, Blau O.1, Kunitz A.1, Pezzutto A.1, Dörken B.1, Blau I.W.1 Charité Universitätsmedizin Berlin, Department of Hematology, Oncology and Tumourimmunology, Berlin, Germany

Introduction: Interaction of multiple myeloma (MM) cells with bone marrow stromal cells (BMMSCs) is essential for their proliferation and survival. MM cells utilize glycolytic pathways even in the presence of oxygen leading to increased production of lactate. Lactate is exchanged between cells by monocarboxylate transporters as MCT1 and MCT4. We aimed to investigate the role of MCT1 and MCT4 for the interaction between BMMSCs and MM cells. Methods: BMMSCs from MM patients (MM-BMMSCs; n = 30) were isolated and cultivated as previously described. Co-cultures were performed with KMS12-PE, OPM-2 and JJN-3 cells. mRNA levels were quantified using Real-Time PCR. Protein levels were detected by western blot. CD147 expression was measured using flow cytometry. Lactate was estimated using the “Lactate Assay Kit” (Sigma-Aldrich) and NF-κB activity was detected using the “NF-κB p50/p65 Transcription Factor Assay Kit” (Abcam). Apoptosis of MM cells was measured using Annexin-V. Results: Co-cultivation with MM cells induced up-regulation of MCT1 in MM-BMMSCs (p < 0.042). MCT4 was up-regulated in all co-cultured MM cell lines (p < 0.04). Gene expression changes on mRNA level were reproducible on protein level. In addition, the assumed chaperone of MCT1 and MCT4, CD147, was up-regulated in co-cultured MM cell lines (p < 0.015). Significant up-regulation of CD147 was also seen in MMBMMSCs co-cultured with JJN-3 cells (p = 0.0474). Lactate levels were analyzed in 27 cell culture supernatants. In 18 samples a 2-fold increase was detected (p < 0.0001) whereas 9 samples showed 2-fold reduction of lactate quantity (p = 0.0039). Treatment of co-cultures with MCT inhibitor α-cyano-4-hydroxycinnamic acid (α-CN) led to significant reduction of living KMS12-PE and OPM-2 cells (12% to 24%; p < 0.04). In contrast, α-CN did not induce apoptosis or death in MM-BMMSCs. However, inhibition of MCT transporters was sufficient to inhibit p50 and p65 DNA binding degree up-regulation in co-cultured MM-BMMSCs (p < 0.05). Conclusions: Interaction between MM cells and MM-BMMSCs leads to up-regulation of monocarboxylate transporters MCT1 and MCT4 and to the exchange of lactate. Inhibition of MCTs induces apoptosis of MM cells and inhibits up-regulation of NF-κB activity in MM-BMMSCs. Further exploration of the metabolic interplay between MM cells and MMBMMSCs could provide new starting points for anti-myeloma treatment, for example by inhibition of lactate exchange using MCT inhibitors. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

233


CONTENTS AUTHOR INDEX

P772

Impact of Lenalidomide on T cell subset composition and PD-1 phenotype in patients with multiple myeloma Danhof S.1, Schreder M.1, Knop S.1, Gogishvili T.1, Einsele H.1, Hudecek M.1 Universitätsklinikum Würzburg; II. Medizinische Klinik, Hämatologie und medizinische Onkologie, Würzburg, Germany 1

Introduction: Lenalidomide (Len) has become an integral component of multiple myeloma (MM) therapy and has tumoricidal and immunomodulatory effects, the latter are of particular interest in long-term therapy. However, the precise mode of action and consequences of Len on T cell composition and function are still incompletely understood. Here, we analyzed the distribution of CD8+ and CD4+, naïve and memory T cell subsets in 4 MM patient cohorts including patients before, during and after exposure to Len, and the influence of Len therapy on expression of PD-1. Methods: We performed multi-parameter FACS analysis on peripheral blood of 44 MM patients to identify CD4 and CD8 naïve (TN, 45RA+62L+), central (TCM, 45RO+62L+) and effector (TEM, 45RO+62L-) memory T cells and their PD-1 expression. Analysis was performed in 4 clinical subgroups at primary diagnosis (PDX, n = 12), relapse (RD, 10), during Len maintenance (LenMT, 14) and in sustained complete remission without recent anti-myeloma therapy (sCR, 8). Results: CD4+ T cell counts were within normal range at PDX, but reduced (p < 0.003) in all other cohorts, with lowest counts in RD patients. In contrast, CD8+ counts were normal and comparable in all subgroups, resulting in a lower CD4/CD8 ratio in all patients who had previously undergone myeloma therapy. CD4 subset analysis showed stable TCM counts in patients receiving LenMT, whereas TCM counts were significantly lower in patients with RD and sCR compared to PDX or LenMT (p < 0.008). Within the CD8 subset, a reduction in absolute cell counts was found for CD8 TN in LenMT patients, leading to a lower naïve to memory ratio. PD-1 was detectable on all CD8+ and CD4+ T cell subsets irrespective of treatment state. Within the CD8+ T cell compartment, PD-1 expression was increased in active disease (PDX, RD) as compared to Len MT or sCR. However, no significant difference was found when comparing PD-1 levels of the patient subgroup in complete remission receiving Len MT to untreated patients in sCR. Conclusion: Our data show that LenMT modulates T cell subset composition in MM patients, underlining its potent immunomodulatory capacity. As T cell subsets differ in intrinsic programming and function, our data may guide the integration of novel T cell based therapies into MM treatment. Finally, our data of low PD-1 expression associated with LenMT suggest the potential to use PD-1 check-point blockade for MM therapy to further improve outcome. Disclosure: Sophia Danhof: Other Financial Relationships: travel grant (Celgene GmbH). Michael Hudecek: No conflict of interest disclosed. P773

The role of Gas6-Mer axis in multiple myeloma Waizenegger J.S.1, Ben Batalla I.1, Weinhold N.2, Meissner T.3, Wroblewski M.1, Janning M.1, Riecken K.1, Binder M.1, Atanackovic D.4, Taipaleenmäki H.1, Schewe D.5, Sawall S.1, Gensch V.1, Cubas Cordova M.1, Seckinger A.2, Fiedler W.1, Hesse E.1, Kröger N.1, Fehse B.1, Hose D.2, Klein B.6, Raab M.S.2, Pantel K.1, Bokemeyer C.1, Loges S.1 Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany, Universitätsklinikum, Heidelberg, Germany, 3The Scripps Research Institute, La Jolla, United States, 4University of Utah, Salt Lake City, United States, 5 Universitätsklinikum Schleswig-Holstein, Kiel, Germany, 6CHU Montpellier, Montpellier, France 1 2

Multiple Myeloma (MM) is the second most common hematological malignancy today. Although therapy has been improved, only 30% of patients survive >10 years. Therefore development of novel therapeutic strategies is warranted. Growth-arrest specific gene (Gas6), a Vitamin K dependent

234

Oncol Res Treat 2015;38(suppl 5):1–270

protein, and its receptors Tyro3, Axl & Mer (TAMR) are abundantly expressed in physiological and malignant hematopoiesis. Interestingly, Gas6 can be secreted by the bone marrow stroma and promotes proliferation and survival of malignant cells. Furthermore, it can activate osteoclasts. Therefore we set off to study the role of Gas6 and its receptors in MM and its microenvironment. Expression of Gas6 & TAMR in MM patients and healthy controls was performed using RT-PCR, Western Blot, ELISA and FACS. Overexpression and silencing of Gas6 and TAMR were achieved by lentiviral gene transfer. Co-cultures of MM cell lines with murine stromal cells were carried out. Human Gas6 or murine Gas6 levels were assessed using ELISA. To model MM in vivo we utilized the orthotopic mouse model U266. Expression analyses indicated increased Gas6 and Mer expression in malignant plasma cells, MNCs and bone marrow plasma (BMP) from MM patients compared to healthy donors (11.3 ± 3.4 ng/ml vs. 3.8 ± 0.5 ng/ ml in BMP; n = 7/17; *P < 0.05 and data not shown). In contrast, Axl and Sky were not differentially expressed. To investigate the functional impact of Gas6 & TAMR silencing of Gas6 and Mer in vitro was performed and decreased growth of different MM cells (n = 3; *P < 0.05). Lentiviral silencing of Mer and Gas6 in the U266 model led to decreased MM burden (n = 4/4, *P < 0.05;) and increased overall survival (n = 4/5, *P < 0.05) and. Vitamin K blockade by Warfarin yielded similar results in vivo and reduced cell growth in vitro. Co-cultures using human MM cells and murine stromal cell lines or primary murine mesenchymal stromal cells (MSCs) indicated upregulation of murine Gas6 by OP9 cells and MSCs in comparison to single culture (OP9 >10 fold; MSCs >2 fold; *P < 0.05). Interestingly Gas6 upregulation was impaired when direct cell contact was impeded by cell culture inserts. Notably human MM-derived Gas6 levels remained unchanged. Thus MM cells induce increased Gas6 secretion by cells of the MM microenvironment, which could further stimulate MM cells and osteoclasts. Gas6 & Mer foster growth of MM cells by autocrine and paracrine interactions and represent novel therapeutic targets in this disease. Disclosure: No conflict of interest disclosed. P774

Results of the interdisciplinary Comprehensive Cancer Center Freiburg (CCCF) Tumor Board (TB)-´Multiple Myeloma´ (MM) in terms of TB-recommendations, patient (pt) outcome, inclusion of difficult-to-treat-MM pts in clinical trials (CT) and satisfaction of patients, participants and referring physicians Selder R.M.1, Möller M.-D.1, Pandurevic M.1, Waldschmidt J.1, Ihorst G.2, Herget G.W.3, Henne K.4, Hauschild O.3, May A.M.5, Pantic M.1, Duyster J.1,6, Vach W.7, Wäsch R.1,6, Engelhardt M.1,6 University of Freiburg (UKF) Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 2University of Freiburg (UKF) Medical Center, Clinical Trials Unit, Freiburg, Germany, 3University of Freiburg (UKF) Medical Center, Clinic of Orthopaedics and Traumatology, Freiburg, Germany, 4University of Freiburg (UKF) Medical Center, Radiation Oncology, Freiburg, Germany, 5University of Freiburg (UKF) Medical Center, Institute of Clinical Pathology, Freiburg, Germany, 6University of Freiburg (UKF) Medical Center, Comprehensive Cancer Center Freiburg – CCCF, Freiburg, Germany, 7University of Freiburg (UKF) Medical Center, Clinical Epidemiology, Freiburg, Germany 1

Introduction: TBs are essential in modern oncology to provide state-ofthe art therapies regarding clinical pathways (CP) and interdisciplinary pt care. Our weekly-held MM-TB started in 2012 with participating hematologist-oncologists, orthopaedists, radiotherapists, pathologists, cytogenetic specialists, radiologists and, if necessary, others. After the conference, TB-recommendations are available within our electronic pt information system. Methods: This analysis assessed a) TB-recommendations b) frequency of difficult-to-treat MM pts to include in CT, c) satisfaction of pts, participants and referring physicians via standardized questionnaires and d) PFS/OS of MM-TB pts compared to the literature (Kumar, Leukemia 2012). TB-advice was based on CP and up-to-date literature. Using the

Abstracts


CONTENTS AUTHOR INDEX

GRADE criteria, all TB-decisions and their level of evidence were assessed (Engelhardt, Haematologica 2014). Results: From 6/2012 to 6/2014, 498 TB-decisions were assessable. 81% TB-presentations involved MM pts, but pts with MGUS, SMM, rare entities/uncertain diagnosis were also discussed. TB-discussions substantially involved relapsed and/or refractory pts. All recommendations were consistent with CP or international literature. 78% TB-decisions were assigned to level of evidence grades 1A-1C and 22% to grades 2A-2C. Importantly, 94% MM-TB decisions were followed. Of 288 pts discussed within the TB, 15% could be successfully included in CT. Detailed analyses of questionnaires of participants, referring physicians and pts revealed gratifying results: 58 participants acknowledged the TB as a useful institution for best possible pt care and their benefits in joining the TB. Of 30 interviewed referring physicians, all were fully satisfied both with the proposed diagnostics and therapies. Notable, however, 17% reported some loss of information, which displays the relevance of the constant and intense interdisciplinary interaction with referring colleagues. Of 100 interviewed pts, all considered their care ideally achieved by the TB, 98% stated that their local physicians profited from the TB and 99% responded that their preferences were respected. Conclusions: Our results demonstrate that the MM-TB is a highly relevant platform which facilitates interdisciplinary state-of-the-art cancer care, especially for challenging-to-treat pts. Moreover, it allows physicians to efficiently cooperate and is highly appreciated by cancer pts.

one could determine CS, given that the pt is alive and progression-free or alive, but has progression at time s (Zamboni. JCO 2010). Analysis of the above additional variables from diagnosis to prediction time s may refine CS towards an even more specifically determined prognosis.

Disclosure: Ricarda Selder: No conflict of interest disclosed. Monika Engelhardt: Expert Testimony: Educational Grant Celgene.

Zober A.1, Hieke S.2, Ihorst G.3, Pantic M.1, Duyster J.1, Schumacher M.2, Wäsch R.1, Engelhardt M.1

Fig. 1. Disclosure: No conflict of interest disclosed. P776

Prospective assessment of a comorbidity and functional geriatric assessment (CF-GA), including the revised Freiburg Comorbidity Index (rFCI) in consecutive elderly MM patients (pts)

University of Freiburg Medical Center, Department of Hematology and Oncology, Freiburg, Germany, 2Institute for Medical Biometry and Statistics, University Medical Center, Freiburg, Germany, 3Center of Clinical Trials (ZKS), Freiburg, Germany 1

P775

Conditional survival (CS) in Multiple Myeloma (MM) provides detailed prognostic information of outcome in a large MM data set

Background: Prognosis is usually expressed as survival probability. Overall survival (OS), however, does not reflect, how prognosis evolves over time and is not very informative for patients (pts) who already survived 1 or more years. Conditional survival (CS) describes probabilities of surviving t additional years given they survived s years, provides relevant information, how prognosis changes over time and is the simplest form of a dynamic prediction. We illustrate the use of CS in a large cohort of MM pts with long-term survival which is mandatory for the calculation of CS (Hieke, ... Engelhardt, Schumacher. CCR 2015). Methods: We analyzed 816 MM pts treated at our department between 1997–2011 and assessed 21 variables including gender, age, stage and admission period. We calculated 5-years CS (5y-CS) and stratified 5yCS according to disease- and host-related risks. Component-wise likelihood-based boosting and variables selected by boosting were investigated in a multivariable Cox model. Results: The 5-/10-y-OS were 50% and 25%, respectively. The 5y-CS remained stable (~50% even at 5y; Fig. 1A). Gender did not, but age and advanced D&S stage effected CS (25% for pts≥70y vs. 65% for < 60y, 75% vs. 42% by D&S stage I vs. II+III). Multivariate survival analysis via Cox proportional hazard model determined D&S stage II+III, age >70y, hemoglobin <10 g/dl, ß2-MG ≥5.5 mg/dl, LDH ≥200 U/l as significant risks. Renal impairment, low albumin and unfavorable cytogenetics increased the risk, but failed to reach significance (Fig. 1B). Cytogenetics, response, response duration and other risk parameters post treatment are currently included in our assessment. Conclusions: CS is a starting point for identifying highly relevant factors related to long-term survival. We defined CS by using the fact that the pt is alive at the prediction time s as the conditioning event. Alternatively,

Introduction: Older cancer pts present with a highly heterogeneous health status and treatment choices are often numerous. Therefore, careful assessment of individuals´ condition is important in the execution of oncologic care. In order to define the best treatment for older pts, novel parameters and metrics for non-disease variables are needed. Albeit impairment in Activities of Daily Living (e.g. IADL), Karnofsky Performance Status (KPS) and quality of life (QoL) are predictive for outcome in MM, the prognostic variables within a battery of established functional tests has rarely been delineated nor their combination with disease-related risk factors and molecular markers. Methods: We prospectively performed a comorbidity and functional geriatric assessment (CF-GA) in consecutive MM pts treated at our center according to our institutional CCCF-pathway. This CF-GA included the IADL, Timed Up and Go-Test (TUGT), malnutrition, pain, rating of fitness, SF12-QoL and geriatric depression scale (GDS). Moreover, different comorbidity (CM) scores, namely ß2MG/eGFR (...Engelhardt. EJH 2009), initial Freiburg Comorbidity Index (iFCI), revised FCI (rFCI), Kaplan Feinstein (KF), HCT-CI and CCI were assessed. Results: Characteristics of 119 prospective pts, currently included in this CF-GA, were typical for tertiary centers with a median age of 63 years (range 21–93), mostly advanced disease, median hemoglobin of 10.9 g/ dl (7.6–14.7), eGFR of 70 ml/min/1.73 qm (7–123), ß2-MG of 4.4 mg/l (1.6–38.4) and bone marrow infiltration of 45% (3–90). The baseline frailty assessment revealed a median KPS of 80% (40–100), fitness of 4 (1–6) and functional results for the IADL of 5 (1–8), pain of 2 (0–10), malnutrition of 4 (0–14), MMSE (cognitive deficiency) of 28 (16–30), GDS of 3 (0–13) and TUGT of 10 (4–30). Median CM scores were substantially different with an iFCI of 0, ß2MG/eGFR of 1, KF of 1, HCT-CI of 2, rFCI of 4 and CCI of 7. Therefore, highly valuable and relevant CF-GA-tools seem currently the IADL, TUGT and rFCI. Conclusions: Our CF-GA and rFCI contain easily assessable and reliable tests, that are of interest to further assess for their discriminative character in subgroups of MM pts. Moreover, most predictive CF-CA tools need to be determined in prospective multicentre cohorts and are of interest to include in future trials. We advocate our CF-GA and rFCI to foresee treatment toxicity, facilitate treatment decisions and guide personalized therapies.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Promny I.1, Hieke S.2, Schinke M.1, Waldschmidt J.1, Ihorst G.3, Pantic M.1, Duyster J.1, Wäsch R.1, Schumacher M.2, Engelhardt M.1 University of Freiburg Medical Center, Department of Hematology and Oncology, Freiburg, Germany, 2Institute of Medical Biometry and Medical Informatics, University Medical Center, Freiburg, Germany, 3Clinical Trials Unit, University Medical Center, Freiburg, Germany 1

235


CONTENTS AUTHOR INDEX

Disclosure: Alexander Zober: Expert Testimony: Deutsche Krebshilfe grant 111424 (to ME+RW). Monika Engelhardt: Expert Testimony: Deutsche Krebshilfe grant 111424 (to ME+RW).

Danhof S.1, Schreder M.1, Rasche L.1, Strifler S.1, Steigerwald A.2, Einsele H.1, Knop S.1

P777

Practice patterns of multiple myeloma management in Germany

Universitätsklinikum Würzburg; II. Medizinische Klinik, Hämatologie und medizinische Onkologie, Würzburg, Germany, 2Universitätsklinikum Würzburg, Zytostatika-Abteilung, Würzburg, Germany 1

Raab M.S.1, Schöhl M.2, Ikenberg R.2 Universitätsklinikum, Heidelberg, Germany, 2Amgen GmbH, München, Germany 1

Introduction: Multiple myeloma (MM) management has significantly evolved in Germany. However, limited data on real-life patient management in clinical practice are available. The aim of this study is to investigate treatment patterns and patient characteristics in symptomatic MM in Germany. Methods: Data were collected using detailed retrospective medical records by oncologists/haematologists managing patients with MM. Patient treatment patterns and characteristics of patients seen in the previous 3 months were analysed, with a quota for patients who had completed specific treatment lines. Results: A total of 1,190 patient charts were reviewed by 100 physicians. 35% of physicians were office-based, 26% in university hospitals and the remaining in cancer centers and other hospitals. At diagnosis, 65% of patients had bone pain and 35% anemia. 55% of patients had ≥1 negative prognostic factor (e.g. ≥2 bone lesions, haemoglobin < 8.5g/dL). Only few patients showed a history of neuropathy (3%) or deep vein thrombosis (7%) prior to the MM diagnosis. Bortezomib based regimens were most widely used in 1st line (1L) (81%) and lenalidomide based regimens in 2L (60%). For 3L and 4L+, the treatment regimens were more diversified (Table). The sequencing of patients receiving bortezomib in 1L revealed that the majority switched to lenalidomide based regimen in 2L (59%) and the remaining patients were re-treated (18%) or received bendamustine based regimens (11%). Main reason for ending treatment was remission with 63% in 1L and around 40% in 2L, a pre-planned treatment stop was second most common reason. Median duration of treatment lines was declining with line of therapy: 12 months in 1L (5 months on treatment followed by treatment-free interval of 7 m), 10 m in 2L (6m / 4m), and 5 m in 3L (no split). Tab. 1. Anti-tumor drug usage by line of therapy

1st Line n = 411

2nd Line n = 353

3rd Line n = 317

4th Line n = 72

4th Line+ n = 109

Bortezomib

81%

28%

25%

25%

23%

Lenalidomide

2%

60%

27%

12%

14%

Thalidomide

4%

4%

6%

12%

11%

Bendamustine

8%

8%

27%

32%

32%

Pomalidomid

0%

0%

13%

16%

12%

P778

Efficacy and safety of pre-approval carfilzomib-based therapy in multiple myeloma – a “real life” experience from an academic center

Introduction: Bortezomib (bort), the first-in-class proteasome inhibitor (PI) has long been integral part of the standard-of-care treatment in multiple myeloma (MM). Carfilzomib (carf), the second generation PI, was granted approval by the U.S. Food and Drug Administration in 2012, however, the drug has still not been approved by the European health authorities. We here show efficacy and safety data from a “real life” experience at a German academic center, indicating a valid role of carf in combination treatment of advanced MM. Methods: 20 patients were registered within the European Carfilzomib Access Program and treated with a three-drug combination of carf (day 1,2; 8,9; 15,16; starting dose: 20 mg/m², target dose: 27 mg/m²), low dose dexamethasone and either lenalidomide (len, 6 patients), cyclophosphamide (cyclo, 12 patients) or doxorubicin (doxo, 2 patients), respectively. Individual decisions were made based on each patient’s treatment history and organ functions. Patients received first treatment with carf at a median of 72 months from primary diagnosis and after a median of 6 previous lines of therapy. All patients had previously been treated with bort and immunomodulatory drugs, with 17/20 (85%) patients refractory to bort and 19/20 (95%) refractory to len or pomalidomide. Results: 18/20 (90%) patients completed at least 1 treatment cycle and were thus eligible for response evaluation. They had received a median of 4 treatment cycles. Overall response rate (ORR) in this heavily pre-treated patient group was 61% with a median duration of response of 6.8 months. Median progression free survival (PFS) was 6.0 months and median overall survival was 6.6 months. Analyzing outcomes with regards to the combination partner, use of cyclo resulted in at least comparable ORR (67% vs. 50%) and PFS (6.8 vs. 2.4 months) as opposed to len. Grade 3/4 adverse events (AEs) occurred in 10 patients (50%), including lung or device related infections (both 10%), febrile neutropenia (15%) and left ventricular failure (LVF, 25%). We found that previous radiotherapy of the thoracic spine increased the risk of cardiac AEs from 17% to 50%. Furthermore, both patients who had received doxo in combination with carf developed acute LVF requiring intensive care treatment. Conclusion: Carf-based combination therapy is effective in extensively pre-treated MM patients. However, we found an increased risk of severe cardiac AEs, most likely due to various cumulative toxicities. Disclosure: Sophia Danhof: No conflict of interest disclosed. Stefan Knop: Advisory Role: Onyx Pharmaceuticals Inc., Amgen GmbH; ­Financing of Scientific Research: Amgen GmbH.

Conclusion: Whereas clear treatment patterns are observed for patients in 1L and even 2L, those patients who reached later lines are treated with a wide range of therapies. These real-world data suggest that there is a strong medical need for innovative therapies in Germany. Disclosure: Marc Raab: Advisory Role: Amgen, Celgene , Novartis; Expert Testimony: Novartis, Morphosys. Robert Ikenberg: Employment or Leadership Position: Mitarbeiter Amgen GmbH; Stock Ownership: Aktien Amgen GmbH.

236

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

P779

P780

A retrospective multicenter analysis of clinical care and outcome in 178 patients with newly diagnosed symptomatic multiple myeloma in Berlin and the area of Brandenburg, between 2008 and 2011

High-dose chemotherapy followed by autologous stem cell transplantation in elderly patients with multiple myeloma – a monocentric retrospective study on effectiveness and safety

Jakob C.1, Kiewe P.2, Pohlkamp C.3, Oldenkott B.3,4, Kingreen D.5, Hopfer O.6, Jehn C.7, Linde H.8, Lüder F.9,10, Jahnke K.10, Göner M.11, Josting A.12, Sallmann D.13, Rothmann F.1, Maschmeyer G.1, Derwahl K.-M.3 Klinikum Ernst von Bergmann, Klinik für Hämatologie, Onkologie und Palliativmedizin, Potsdam, Germany, 2Onkologischer Schwerpunkt am Oskar-Helene-Heim, Berlin, Germany, 3St. Hedwig-Krankenhaus, Klinik für Innere Medizin, Berlin, Germany, 4Poliklinik Große Hamburger-Str., MVZ für Innere Medizin, Hämatologie/Onkologie, Berlin, Germany, 5Onkologische Schwerpunktpraxis Tiergarten, Berlin, Germany, 6Klinikum Frankfurt/Oder, Medizinische Klinik I, Frankfurt/Oder, Germany, 7Charité – Universitätsmedizin Berlin, Med. Klinik m.S. Hämatologie, Onkologie, Tumorimmunologie, Berlin, Germany, 8MVZ für Blut- und Krebserkrankungen, Potsdam, Germany, 9Klinikum Brandenburg, Zentrum Innere Medizin II, Abteilung Onkologie/Palliativmedizin, Brandenburg, Germany, 10Onkologische Schwerpunktpraxis, Brandenburg, Germany, 11MVZ am St. Josefs-Krankenhaus, Potsdam, Germany, 12Onko Berlin, Berlin, Germany, 13Ruppiner Kliniken GmbH, Medizinische Klinik B, Neuruppin, Germany 1

Introduction: Over the last two decades, there was substantial improvement in survival of patients diagnosed with multiple myeloma (MM). Important milestones were the implementation of high-dose therapy (HDT) with autologous stem cell transplantation (ASCT) and the introduction of new drugs. Despite the improvement in remission rates and overall survival (OS) in clinical trials, epidemiological studies suggest a greater benefit in younger patients (≤65 yrs), while only little improvement in elderly patients or patients with comorbidities, who are both underrepresented in clinical trials. Methods: To evaluate the practical implementation of novel diagnostic and therapeutic strategies in the routine care of MM, we reviewed disease courses and outcomes of 178 consecutive patients with newly diagnosed symptomatic MM, referred to 13 cancer centers between 2008 and 2011 in the metropolitan area of Berlin and the region of the state of Brandenburg. Results: In academic institutions or large cancer centers there was a higher proportion of patients ≤65 yrs and with ECOG-PS <2 (68 and 85%) compared to regular care institutions (60 and 31%, respectively). Baseline cytogenetic analysis was available in 81% in younger and in 50% in elderly patients and was performed more frequently in academic versus non-academic institutions (84% vs. 50%). The proportion of patients in clinical trials was twofold higher in academic institutions (44 vs. 21%). First-line bortezomib was administered at 65% in patients ≤65 yrs and 29% in >65 yrs. Tandem HDT/ASCT in patients ≤65 yrs and HDT in patients 65–70 yrs were performed more frequently in patients with primary diagnosis at a transplant center compared to those diagnosed in centers without a transplantation unit (42 vs. 28% and 72 vs. 40%, respectively). Overall survival at 2 yrs was 71% in patients ≤65 yrs and 61% in elderly patients. There were no significant differences in OS between patients treated in large cancer centers versus those treated in community based hospitals or private practice. Conclusion: Our analysis shows a consistently high standard of care in terms of adherence to current guidelines and availability of novel treatments. The higher rates of HDT/ASCT and first-line administration of new drugs in academic institutions or cancer centers can be attributed to the higher proportions of younger patients and patients in clinical trials in those centers. Disclosure: No conflict of interest disclosed.

Tzalavras A.1, Kasenda B.1, Illerhaus G.1, Schleicher J.1 Klinikum Stuttgart, Katharinenhospital, Stuttgart, Germany

1

Background: High-dose chemotherapy followed by autologous stem cell transplantation (HDT-ASCT) is considered standard treatment for eligible patients <65 years with multiple myeloma (MM). However, most patients are older than 65 some of which may also be eligible for HDT-ASCT. Therefore the cut-off at 65 years is probably not appropriate anymore in an aging patient population. We sought to investigate effectiveness and safety in elderly MM patients who were treated with HDT-ASCT. Patients and methods: We conducted a retrospective analysis of consecutive MM patients ≥60 years being treated with HDT-ASCT at the Klinikum Stuttgart (Germany) between 1999 and 2014. Main safety outcomes included occurrence of treatment related infectious complications, transfusion rates and treatment associated mortality. Main outcomes for effectiveness included progression free survival (PFS) and overall survival (OS). Results: We analyzed 62 patients, 42 (67.7%) were male. The median age at HDT-ASCT was 66.4 years. 23 (37.1%) were 60–64, 23 (37.1%) 65–69 and 16 (25.8%) ≥70 years. All patients had a good performance status (Karnofsky-Index ≥80). Disease status before HDT-ASCT: 28 (45.2%) were in partial remission or very good partial remission, 15 (24.2%) had progressive disease, 5 (8.0%) had a relapse and 6 (9.7%) a stable disease. In 7 (12.9%), disease status before HDT-ASCT was not available. All patients were treated with high-dose melphalan (MEL): 18 (29.0%) with MEL-200 mg/m2, 38 (61.3%) with MEL-140 mg/m2, 3 (4.8%) with MEL-100 mg/m2 and 3 with MEL-70 mg/m2. All patients received peripheral blood stemcells. Overall, the median absolute neutrophil-count recovery time was 10.6 days; 10.7 for patients 60–64 years, 10 days for those 65–69 years and 11.3 days for patients ≥70 years. No patient died during or in the first 100 days after HDT-ASCT. Final results of our analysis will be presented at the meeting. Conclusion: HDT-ASCT in elderly MM patients is feasible in selected patients and treated related mortality does not seem to be higher if patients are well selected. More detailed results will be presented at the meeting. Disclosure: No conflict of interest disclosed.

Posterdiskussion Multiples Myelom II P781

Post allogeneic transplant antibodies target epitopes exposed on the cell surface after bortezomib treatment in multiple myeloma Schieferdecker A.1, Oberle A.1, Thiele B.1, Braig F.1, Düsedau A.2, Miethe S.3, Hust M.3, Bacher U.4, Kröger N.5, Binder M.1 University Medical Center Hamburg-Eppendorf, Department of Oncology and Hematology, BMT with section Pneumology, Hubertus Wald Tumorzentrum / UCCH, Hamburg, Germany, 2Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Virus Immunology, Hamburg, Germany, 3Institute for Biochemistry and Biotechnology, Department of Biotechnology, TU Braunschweig, Braunschweig, Germany, 4University Medical Center Göttingen, Department of Hematology and Medical Oncology, Göttingen, Germany, 5 University Medical Center Hamburg-Eppendorf, Department of Stem Cell Transplantation, Hamburg, Germany 1

Introduction: After allogeneic transplant myeloma patients may develop humoral immune responses against proteins overexpressed in myeloma cells. This subset of patients seems to have a favorable prognosis. However, the events triggering such antibody formation as well as their functional role remain unclear.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

237


CONTENTS AUTHOR INDEX

Methods: Epitope mimics targeted by post-transplant sera were identified by random peptide phage display screenings. Mimics were traced back to their parental antigen by reverse immunization and proteomic methods. Immune repertoires were produced as scFv phage libraries and interrogated for cell membrane exposed antigens by cell-based selection and next-generation sequencing. Cell translocation of HSP71 with bortezomib treatment was analyzed by flow cytometry. Results: We characterized a landscape of epitopes targeted by myeloma patient immune responses in the post-transplant setting. These epitopes were highly myeloma-specific since they were not recognized by sera from healthy donors or patients transplanted with myeloid malignancies. We argued that these epitopes may only be targets of a direct functional anti-myeloma immune response if exposed on the myeloma cell surface. To test this, we identified patients with highly epitope-reactive sera and used their B- and plasma cells from blood and bone marrow for the construction of phage-displayed scFv antibody libraries. Screening of these libraries on myeloma cell lines and primary myeloma cells, however, did not yield cell surface interacting scFv antibodies, if no cell surface binding control scFv antibodies were spiked in. In line with this, tracing back two of the commonly recognized epitope mimics revealed cytoplasmic proteins (HSP60 and HSP71) as parental antigens. To investigate how these primarily cytoplasmic proteins may trigger humoral immune responses, we investigated membrane translocation of HSP71 after anti-myeloma treatment. Interestingly, we found only very weak membrane exposure at baseline, whilst bortezomib triggered pronounced translocation of HSP71 to the cell surface. Conclusion: Humoral immune responses in patients after allogeneic transplant may be directed against myeloma-specific epitopes exposed on the myeloma cell surface as a result of anti-myeloma treatment. The favorable prognosis of these myeloma patients may be most likely explained by secondary, e.g. T-cell mediated, effects which need to be further studied. Disclosure: No conflict of interest disclosed. P782

Hospital population screening reveals overrepresentation of CD5- monoclonal B-cell lymphocytosis and monoclonal gammopathy of undetermined significance of IgM type Voigtländer M.1, Vogler B.1,2, Trepel M.3, Panse J.4, Jung R.5, Bokemeyer C.1, Bacher U.6, Binder M.1 Universitätsklinikum Hamburg-Eppendorf, II. Med. Klinik und Poliklinik, Hamburg, Germany, 2Sana Klinikum Elmshorn, Abteilung für Anästhesiologie und Intensivmedizin, Elmshorn, Germany, 3Klinikum Augsburg, II. Med. Klinik, Augsburg, Germany, 4Uniklinik Aachen, Klinik für Hämatologie, Onkologie, Hämostasteologie und Stammzelltransplantation, Aachen, Germany, 5 Universitätsklinikum Hamburg-Eppendorf, Institut für Klinische Chemie und Laboratoriumsmedizin, Hamburg, Germany, 6Universitätsmedizin Göttingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany 1

Introduction: Monoclonal B-cell lymphocytosis (MBL) and monoclonal gammopathy of undetermined significance (MGUS), resulting from clonal expansions of mature B- or plasma cells, are considered precursor conditions of B-cell malignancies. Regarding potential host factors that may facilitate the development of lymphoproliferations, one may speculate about a possible association between these precursor lesions. However, very limited data exist on the co-prevalence of MBL and MGUS. Methods: We investigated the immunophenotypic/monoclonal immunoglobulin (M-protein) features and co-prevalence of MBL and MGUS in a hospital-based cohort of 1,909 non-hematooncological patients by fivecolor flow cytometry and immunofixation, respectively. Results: Of the evaluable cases, 3.8% showed evidence for MBL, while 9.8% were screened positive for M-protein. With six concomitant cases (0.4%), MBL and MGUS were not statistically associated. At least in two of these coincident cases, MBL and MGUS were of different clonal origin since both clones had divergent light chain restriction. CD5- MBL (57.1%) and IgM+ MGUS (24.7%) were strikingly overrepresented compared to population-based screenings and did not progress to overt lym-

238

Oncol Res Treat 2015;38(suppl 5):1–270

phoma or myeloma during the observation period (mean follow-up of 117 weeks or 110 weeks, respectively). Conclusion: Prevalence and phenotypes suggest that a substantial proportion of incidental MBL and MGUS in hospitalized patients may be attributed to transiently expanded B-cell clones in the context of disease-related immune stimulation rather than reflecting veritable precursors of clonal B-cell malignancies. Disclosure: No conflict of interest disclosed. P783

Revlimid-NIS: Non-interventional study on therapy of relapsed/ refractory multiple myeloma with a combination of lenalidomide plus dexamethasone Knauf W.1, Aldaoud A.2, Maintz C.3, Groschek M.4, Teichmann B.5, Harde J.5, Trarbach T.5 Centrum für Hämatologie und Onkologie Bethanien, Frankfurt a. M., Germany, 2Gemeinschaftspraxis Hämatologie/Onkologie, Leipzig, Germany, 3 Hämatologisch-Onkologische Praxis, Würselen, Germany, 4Hämatologie – Onkologie – Stolberg, Stolberg, Germany, 5iOMEDICO AG, Freiburg, Germany 1

Introduction: Multiple myeloma (MM) is the second most common haematological malignancy.The immunomodulatory drug lenalidomide (Len) has demonstrated significant activity in advanced MM. Len in combination with dexamethasone (Dex) is approved for the treatment of patients with MM. Methods: This prospective non-interventional study was initiated to collect data on Len/Dex in routine treatment of patients with MM. Data on demographics, tumour characteristics, comorbidities, and Len treatment were analysed. Stage was determined according to Durie and Salmon. Main endpoints were time to progression (TTP), overall response rate (ORR) and safety parameters. Results: A total of 99 patients were enrolled at 16 sites in Germany between 08/2009 and 04/2012. Of those, 97 were evaluable. Median age was 70.7 (range 42.3–88.1) years, with 63.9% males. At inclusion most patients had an ECOG PS of 0 or 1 (60.9%; n = 59), for n = 37 ECOG PS was not evaluated at baseline. The majority (67%; n = 65)) had stage III disease (according to Durie and Salmon) and 68.1% (n = 66) were diagnosed with heavy chain class IgG. 53.6% had received more than one prior therapy. Renal function was impaired in 24.7% of patients according to investigator’s records. Median TTP was 12.1 months (95% CI 9.2–14.6), ORR (including minor responses) was 60.8% (59 responder, 23 non-responder, with n = 15 missing data). Median OS was 24.3 months (95% CI, 19.5– 33.1). Overall, most common adverse events with suspected relation to Len/Dex were leukopenia (23.7%), thrombocytopenia (22.7%), anaemia (17.5%); fatigue (11.3%) and peripheral neuropathy (11.3%). Thromboembolic events occurred in 5 patients of whom one had not received prophylaxis. 33% (n = 32) of patients had CTC AE grade 3 or 4 events. No therapy related AEs with fatal outcome were reported. Conclusions: Treatment with Len/Dex is effective and well tolerated in patients with relapsed/ refractory MM. No unexpected toxicities were reported. The response rate as well as TTP and OS were comparable to those reported in the registration trial. Disclosure: Wolfgang Knauf: Financing of Scientific Research: Celgene. Tanja Trarbach: Employment or Leadership Position: iOMEDICO AG.

Abstracts


CONTENTS AUTHOR INDEX

P784

Using IgM hevylite to distinguish MGUS from malignant disorders with clonal IgM Paraprotein Scheid C.1, Blommer J.2, Holtick U.1, Chemnitz J.1, Malchau G.2, Streichert T.2, Hallek M.1 Klinik I für Innere Medizin, Uniklinik, Köln, Germany, 2Institut für Klinische Chemie, Uniklinik, Köln, Germany 1

Introduction: Monoclonal IgM (mIgM) can be found in elderly patients without evidence of any haematological disorder but also in patients with lymphomas (NHL), or myeloma (MM). Both show a positive immunofixation but have a quite different prognosis and management. Therefore it is crucial to have diagnostic tools available that can rapidly distinguish these two scenarios. In this analysis the value of using the IgM Hevylite® kappa and lambda assays (IgM-HLC, Binding Site) to distinguish MGUS from lymphoma or myeloma was assessed. Methods: From 04/2011 to 11/2012 aliquots of serum samples taken for routine immunofixation were stored at the Institute for Clinical Chemistry at the University Hospital Cologne. For patients with positive mIgM kappa or lambda immunofixation the IgM-HLC was performed by Binding Site. The analysis was approved by the ethics committee of the University of Cologne. Results: 64 patients were identified with mIgM (39 kappa, 25 lambda) and assessed for IgM-HLC. 3 patients had missing clinical information, therefore 61 patients (36 MGUS, 22 NHL, 3 MM) could be analysed. An abnormal HLC kappa/lambda-ratio (rHLC) was found in 33/36 (91.7%) MGUS and 24/25 (96.0%) NHL/MM patients (not significant). The median rHLC for IgM kappa was 11.1 in MGUS and 142.7 in NHL/MM patients and for IgM lambda 0.54 and 0.03, respectively. Both differences were highly significant (p = 0.001, Mann-Whitney-U test). A suppressed non-involved IgM kappa or lambda was present in 3/36 (8.3%) MGUS and 16/25 (64.0%) NHL/MM patients (p < 0.001, Chi-square-test). Conclusion: While an abnormal IgM-rHLC is present in most cases with a positive IgM immunofixation, both the numerical value of the rHLC and the suppression of the non-involved IgM seem to distinguish malignant from non-malignant conditions presenting with a mIgM. IgM-HLC therefore deserves further investigation in the diagnostic workup of patients with mIgM. Disclosure: Christof Scheid: Financing of Scientific Research: binding site, ­Janssen, Celgene, Amgen. Michael Hallek: No conflict of interest disclosed.

Blood 2015). Therefore, detecting early and especially symptomatic MM is important, albeit MM symptoms are often unspecific and diagnosis finding is proposed to take at least 6 months (ms) or longer. Objective literature is, however, lacking and possible risks which might lead to latencies in diagnosis remain undetermined. Methods: 108 MM pts diagnosed between 1997–2014, treated within clinical trials at Medical Center University of Freiburg and discussed in our MM tumor board, were meticulously analysed by their medical reports. We assessed the duration and type of initial symptoms occuring until MM is diagnosed and whether specific risks for possible delays can be determined. For a further prospective analysis, we developed a MM-specific questionnaire to assess pts seen in our outpt clinic. Results: 102 of 108 retrospective pts had symptoms before the diagnosis of MM, in 16 pts (15%) prior MGUS, SMM or solitary plasmocytoma were documented. The median time from first symptoms to the final MM diagnosis was 3ms, albeit with large variation (0–120). Pts frequencies diagnosed within < 3, 3–6, 7–11 and ≥12ms were 39%, 27%, 14% and 20%, respectively. Comparison of initial symptoms in pts diagnosed within ≤6 (n = 68) vs. >6ms (n = 34) did not reveal substantial differences in CRAB-related symptoms. Non-CRAB-related symptoms, like B-symptoms or infections, were also indifferent but more frequent with 35% and 25%, respectively, than previously reported (Engelhardt, Haematologica 2014). Pts diagnosed within >6ms showed increased frequency of light-chainonly MM, prior orthopedic or rheumatic comorbidities, residency in smaller communities and longer distance to tertiary centers compared to pts diagnosed within ≤6ms. Conclusions: Most pts in our cohort were diagnosed within < 3ms from the first symptoms. However, delayed (≥12ms) diagnosis finding was observed in 20%. Differences of initial symptoms in pts diagnosed within ≤vs. >6ms were not significant, whereas others, e.g. disease dynamics and comorbidities, seem more important, warranting further prospective analyses which are currently under way and will be reported at the meeting. Disclosure: Giulia Graziani: No conflict of interest disclosed. Monika Engelhardt: Expert Testimony: Educational Grant: Janssen-Cilag GmbH. P786

Using the Sleeping Beauty transposon system to generate stably transfected myeloma cells for molecular analyses Roth B.1, Fink S.1, Zugelder L.1, Steinbrunn T.1, Chatterjee M.1, Einsele H.1, Bargou R.1, Stühmer T.1 Universitätsklinikum, Würzburg, Germany

1

P785

Are we at our best to avoid delays in diagnosis finding: Assessment of time from onset of first symptoms to the final diagnosis of multiple myeloma – possible risks and future solutions Graziani G.1, Waldschmidt J.1, Herget G.W.2,3, Pandurevic M.1, Henne K.4, Selder R.1, May A.M.5, Möller M.1, Ihorst G.6, Thomsen A.4, Reinhardt H.1, Vach W.3,7, Duyster J.1,3, Wäsch R.1, Engelhardt M.1,3 Medical Center University of Freiburg, Department of Medicine I Hematology and Oncology, Freiburg, Germany, 2Medical Center University of Freiburg, Department of Orthopedics and Trauma Surgery, Freiburg, Germany, 3Medical Center University of Freiburg, Comprehensive Cancer Center Freiburg – CCCF, Freiburg, Germany, 4Medical Center University of Freiburg, Department of Radiation Oncology, Freiburg, Germany, 5Medical Center University of Freiburg, Institute of Clinical Pathology, Freiburg, Germany, 6Medical Center University of Freiburg, Clinical Trials Unit, Freiburg, Germany, 7Medical Center University of Freiburg, Center for Medical Biometry and Medical Informatics, Freiburg, Germany 1

Introduction: Multiple myeloma (MM) evolves from precursor diseases MGUS and SMM and has seen major advances in the understanding and management, among them revised disease definitions and recognition of early-defining risks. Early MM diagnosis and treatment initiation of symptomatic patients (pts) have recently been proposed (Rajkumar,

Abstracts

Introduction: The generation of stably transfected multiple myeloma (MM) cells is either inefficient and time-consuming (selection and propagation of single clones after random plasmid integration) or requires virus-based approaches with the potential disadvantages of having to work under S2 conditions and the variability of viral stocks. We have therefore tested the utility of the Sleeping Beauty transposon system to rapidly generate stably transfected MM cells for molecular analyses. Methods: We have used our established electroporation protocols to introduce the Sleeping Beauty transposon/transposase system into MM cells. Selection of transfected cells was achieved with G418 and/or puromycin, and short-hairpin RNA as well as mRNA expression cassettes were used as payloads. Results: We found that bulk numbers of stably transfected cells can easily be generated within 2–3 weeks with commonly used MM cell lines like AMO-1, INA-6, JJN-3, L-363 and MM.1s. Even without further antibiotic selection protein expression (EGFP) remained stable for at least 6 months and shRNA-mediated knockdown (tested with targets in the MEK1/ MEK2/ERK1/ERK2 module) lasted for at least 2–4 months (longer-term assessments are still ongoing). As the Sleeping Beauty system permits incorporation of relatively large payloads we have also successfully tested its application with multiple shRNA expression cassettes. Current efforts are aimed at developing constructs that are inducible using tet-repres-

Oncol Res Treat 2015;38(suppl 5):1–270

239


CONTENTS AUTHOR INDEX

sor-based approaches and the state of affairs will be presented and is open for discussion at the poster. Conclusions: The Sleeping Beauty system is highly useful for the fast generation of stably transfected populations of MM cells for protein expression and RNAi applications. The ease of use, lack of requirements for biosafety level 2 and the high penetrance and stable implementation of the introduced traits make this an excellent tool for molecular analyses of intracellular signalling pathways in MM cells. Disclosure: No conflict of interest disclosed. P787

Metachronous occurrence of multiple myeloma and hepatocellular carcinoma – coincidence or coherence? Strifler S.1, Schreder M.1, Danhof S.1, Einsele H.1, Knop S.1 Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany

1

The occurrence of second primary malignancies in multiple myeloma (MM) is well known, though, there is an ongoing debate about the underlying mechanisms. An association with immunomodulatory substances has been shown. We identified 3 current cases of metachronous occurrence of hepatocellular carcinoma (HCC) in MM patients (pts.) at our institution. In none of them any typical HCC-related risk factors were found. Two pts., both aged 71 at time of MM diagnosis, had been enrolled in a clinical trial. The first one had received 27 cycles lenalidomide/dexamethasone (Rd) with documented CR when diagnosed with multifocal liver lesions during routine ultrasound, proven to be HCC on liver biopsy. The 2nd patient, treated with Rd-induction, tandem autologous transplant and 2 cycles of R-maintenance with subsequent 3rd cyclophosphamide-based ASCT (due to graft failure) with 12 month’s latency was diagnosed with a large focal liver lesion in MM VGPR being the reason for suddenly elevated transaminases. Due to unequivocal findings (MRI, AFP), no biopsy was needed. Both pts. are treated by local procedures now, i.e. transarterial chemoembolisation (TACE) and radioembolization (SIRT), respectively. Presenting with a 4-year history of MM being in CR after tandem ASCT followed by 4 cycles of VCD at first relapse, and preceding papillary thyroid cancer, successfully treated by thyroidectomy and radioiodine ablation in 2004, the 3rd, 60-year old patient was diagnosed with HCC in 2011. Subsequent prostate cancer requiring transurethral resection was detected in 2012. Initial, HCC-directed treatment had been by partial liver resection and sorafenib maintenance, followed by several systemic therapies (temsirolimus, bevacizumab/erlotinib) and local treatment (TACE and SIRT) due to progression. With increasing signs of liver failure, biopsy focusing on cMET over-expression was done with positive result. We offered treatment with the small-molecule cMET-inhibitor tivantinib but it could not be started anymore due to rapid decline. Since cMET overexpression has been described as a mechanism in MM – cell growth and playing a role in development of a variety of solid cancers, this might be the missing link in these cases besides therapy-induced damage. cMET immunostaining is currently performed in the two other specimens. We conclude that development of HCC in MM pts. deserves investigation as cMET might link the two entities’ tumorigenesis. Disclosure: No conflict of interest disclosed. P788

IgM Multiple Myeloma as a rare cause for bleeding diathesis Pecher A.-C.1, Hinterleitner C.1, Kanz L.1, Jaschonek K.1 Eberhard Karls University, Departement of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmology, Tübingen, Germany 1

Introduction: The IgM Multiple Myeloma (MM) only accounts for 0,5% of the cases of MM. The clinical features of IgM MM do overlap with Waldenstrom´s macroglobulinemia (WM) as far as they are related to IgM monoclonal protein, i.e. hyperviscosity symptoms or bleeding dis-

240

Oncol Res Treat 2015;38(suppl 5):1–270

orders. But both diseases can be distinguished by immunhistochemistry, presence of osteolytic lesions or detection of the MYD88-Mutation respectively translocation t(11;14). Distinguishing these two diagnoses is important as the therapeutic approach and prognosis are quite different. Here we report a case of IgM myeloma confirmed by bone marrow biopsy and immuno-phenotype analysis. Case report: A 54 year old man presented in the emergency department with syncope and massive melena after gastroscopy on the day before admission. He complained of loss of efficiency during the last two months as well as paresthesia and cold extremities over the last three weeks. Physical examination was normal, whereas laboratory testing showed a moderate anemia, renal impairment (creatinine 1,4 mg/dl), elevated serum protein with presence of M-protein and elevated IgM (4020 mg/dl). In further analysis cryoglobulins could be detected, which in addition with a decrease in red cell production explained the considerable variations of hemoglobin. The cryoglobulins can also be taken in account for hyperviscosity syndrom i.e. hemorheologic disturbance with acrocyanosis and reported paresthesia. Assays for von Willebrand disease (vWD) showed positive, explaining the unusual bleeding after gastroscopy. The bone marrow aspirate was consistent with the diagnosis of IgM MM, showing 50% infiltration oft the bone marrow with plasma cells. There was no evidence MYD88 mutation but the cells stained strongly positive for t(11;14). A CT scan revealed no osteolytic lesions. The patient was treated with four cycles of cyclophosphamide, bortezomib and dexamethasone, achieving CR. Willebrandfactor normalized (multimeric pattern and CBA/WF Ratio), cryoglobulins and acral perfusion disturbance disappeared. Discussion: This patient with IgM MM had IgM paraproteinemia and presented with hyperviscosity symptoms as well as bleeding caused by acquired vWD. It is important, as outlined, to distinguish IgM MM from WM as the therapeutic approach and prognosis are quite different. Disclosure: No conflict of interest disclosed. P789

Dermatosis toxica and phlebitis – adverse effects in a patient with multiple myeloma after bortezomib administration subcutaneous and intravenous Piribauer M.1, Tomka M.1, Weiss H.1, Siebert F.1 Krankenhaus Barmherziger Brüder, Innere Medizin/Onkologie, St.Veit/Glan, Austria 1

Introduction: Bortezomib (Velcade) is a proteasome inhibitor and mainly used for the treatment of myelo- and lymphoproliferative disorder. Skin complications of bortezomib treatment are frequently described in the literature. We describe a case of a patient who developed a Dermatosis toxica after administration subcutaneous and an extensive phlebitis on the arm after an attempt to give her a dose intravenous. Case report: In 2012, a 77year-old female patient showed the diagnosis of a Multiple myeloma, plasmocytic subtye, IgG-kappa, stadium IA, ISSScore I. At first, no CRAB criteria existed so we did a therapy like wait and watch. Six months later the MRT-scan showed compression fractures of the vertebral body, especially TH 12 and LWS 2. Further, the IgGkappa-level increased and the blood level (decreased hemoglobin and increased calcium) showed a progression of the disease. Treatment with bortezomib (1,3 mg/m2 body surface area, days 1, 4, 8, 11), melphalan (9 mg/m2, days 1–4) and prednisone (100 mg days 1–4) (VMP) was started. Bortezomib was given first subcutaneous (s.c.). After a four-month therapy (two cycles) a complete response (CR) of IgG kappa was observed. Hemoglobin and calcium normalized too. After further two cycles of therapy a heavy erythema abdominal was observed. After full recovery with topical application of metylprednisolon, we administered bortezomib s.c. on the right thigh. After two days, a central erythema with a central necrosis (where bortezomib was injected) was seen surrounded by a livid area. Dermatological consultations showed the diagnosis of Dermatosis toxica (no picture existing). After recovery with methylpredenislon-aceponat and antihistamine we tried an attempt to inject bortezomib intravenous in the left arm now. A few minutes after administration phlebitis was ob-

Abstracts


CONTENTS AUTHOR INDEX

served (picture). Now we stopped systemic therapy due to intolerance to bortezomib. Conclusions: Subcutaneous administration of bortezomib offers similar efficacy as standard intravenous administration. Bortezomib is classified as an irritant because it causes an inflammatory reaction accompanied with iching, burning when it leaks out. Topical treatment with evening primrose oil relieves this symptom. Severe skin reactions like Erythema multiforme, Vasculitis, Sweet syndrome and histiocytoid Sweet syndrome were described in both application forms. The treatmtent option is methylprednisolon and often stopping treatment with bortezomib. Disclosure: No conflict of interest disclosed. P790

Three rare manifestations of extramedullary Myeloma Witte J.1, Marretta L.1, Rothaug W.1, de Wit M.1 Vivantes Klinikum Neukölln, Berlin, Germany

1

Introduction: Extramedullary Myeloma are a rare entity. They mainly involve the upper respiratory tract. Less common sites include the central nervous system, liver, lymph nodes, gastrointestinal tract, testes and skin. We present three case reports of patients with other rare locations of myeloma manifestations which have been treated at our hospital. First Patient: A 67 year old woman in remission of multiple myeloma after treatment with melphalan, prednisone and thalidomide, was admitted with one week lasting dyspnea. Echocardiography showed pleura and pericard effusions. Transesophageal echocardiography revealed a thickened interatrial septum, as well as a tumor filling half of the atrium. After rapid deterioration the patient, who was also suffering from metastasized breast cancer decided against intensive therapy and died under palliatve care. Postmortem a cardiac manifestation of the multiple myeloma was diagnosed. Second Patient: During a routine checkup in a 62 year old male patient with a history of multiple myeloma in complete remission after high dose melphalan treatment with autologous stem cell transplantation, a small mass was palpable at the right lower back. 6 month earlier a spontaneous intra-pelvic bleeding occured under full anticoagulation. A catheter was place and removed after several days. CT scan showed a tumor following the previous catheter channel. A biopsy confirmed a solitary extramedullar manifestation of the multiple myeloma. Third Patient: A healthy 63 year old woman reported to the emergency room with severe pain in the right shoulder. A CT Scan showed a Pancoast tumor of the right apical lung with multiple bone metastases. A cMRI showed cranial bone metastases with infiltration of the Dura Mater. Laboratory results revealed a normochromic normocytic anemia and a thrombocytopenia. Serum-creatinine and protein was slightly elevated. A CT guided biopsy for the suspected lung cancer revealed the diagnosis of multiple myeloma. Conclusion: Extramedullary manifestations of multiple myeloma are often difficult to diagnose. In patients with multiple myeloma in remission any clinical irregularity should raise the suspicion of relapse.

sometimes with necrosis, arthralgia, and polyneuropathies, often combined with glomerulonephritis. Case report: We report a 61 years old male patient who is mentally deficient from birth and lives in a caring home. In December 2013, the patient noted increasingly blue and later black coloring of his acrae especially of his earlobe and nose. Diagnostic workup led to suspected essential cryoglobulinemia. Accordingly, the patient was treated five times with methylprednisolone and cyclophosphamide. However, treatment improved symptoms only slightely. A relapse in July 2014 was treated with rituximab and also did not cause a significant change of the skin symptoms. In the course of disease progressing thrombocytopenia was noted (platelets <50 Gpt/L). Therefore, a bone marrow histology was performed demonstrating 20% malignant plasma cells. The CRAB criteria were not fulfilled and the disease stage was defined as MGUS subtype IgG kappa (differential diagnosis: smouldering myeloma). The working diagnosis was changed to a secondary cryoglobulemic vasculitis accompanying a hematologic systemic disease (MGUS, DD: smouldering myeloma). Subsequently, the patient was twice treated with melphalan 15 mg/sqm. Under this treatment a notable progress of skin symptoms and a pneumonic sepsis occurred. In January 2015, therapy with bortezomib/dexamethasone was initiated. This therapy caused clinical improvement of the skin lesions and blood count parameters. At present, the patient is being weekly treated with a reduced dose of bortezomib as maintenance therapy. Partial remission of skin symptoms and a improved blood count parameters were observed. Significant immunodeficiency with relapsing infections is an ongoing challenge. Conclusions: Cryoglobulemic vasculitides are rare diseases and may occur as manifestation of a hematologic disease. Therefore, extensive diagnostic workup and reevaluations when appropriate are mandatory as the hematologic disease may manifest later in the course of the disease. Therapeutic decisions should be guided by the hematologic disease. Disclosure: No conflict of interest disclosed.

Posterdiskussion Querschnittsthemen II P792

End-of-Life decision-making in patients with advanced cancer – patients’ information needs and treatment goals in the EPAL study Winkler E.1, Jäger E.2, Mumm F.2, Laryionava K.1, Hiddemann W.2, Heußner P.2 Universität Heidelberg, Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany, 2Medizinische Klinik und Poliklinik III, Klinikum Großhadern, LMU, München, Germany 1

Introduction: Cryoglobulemic vasculitides represent immune complex-mediated diseases of small and medium size vessels occurring more frequently in patients with hepatitis C infection, collagenoses or lymphoproliferative disorders. Typical symptoms include purpura of the skin,

Introduction: End-of-life (EOL) decision-making in cancer patients is often surrounded by clinical, ethical and psychological conflicts and is a challenging process for patients, relatives and their medical team. The aim of the EPAL study (Ethics policy for advanced care planning and limiting treatment) is to develop a clinical practice guideline about limiting life-prolonging treatment and to evaluate its impact on medical practice in a clinic for hematology and oncology. A special focus of this data collection was patient involvement, patients’ information needs and whether these needs are met. Methods: EPAL is a prospective, quantitative study conducted at the Department of Haematology and Oncology at the LMU Hospital, Munich. Recruitment for the pre-implementation finishes in June 2015. Eligible were hospitalized cancer patients with limitations of life-prolonging treatment either being discussed or respective decisions already been taken. We have developed and adopted a set of instruments that is completed both from the patient and the corresponding physician and nurse. Instruments for data collection include patient’s information needs regarding advanced cancer situation, patient preferences goals of care and involvement in decision-making.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

Disclosure: No conflict of interest disclosed. P791

Secondary cryoglobulin vasculitis in a case of MGUS subtype IgG kappa: A difficult diagnosis and therapy with many complications. Gläser D.1, Lestin M.1, Görl N.2, Kneitz C.2, Krammer-Steiner B.1 Klinikum Südstadt Rostock, Hämatologie/Onkologie/Palliativmedizin, Rostock, Germany, 2Klinikum Südstadt Rostock, Rheumatologie/Pneumologie/Geriatrie, Rostock, Germany 1

241


CONTENTS AUTHOR INDEX

Results: Results of the first 50 patient-physician documentation sets will be presented in October 2015 including the proportion of patients involved in the decision process, patient information needs and whether they were met as well as the concordance of patients’ and physicians treatment goals. First trends show that patients report high information needs about diagnosis, treatment options and side effect. Patients are less interested in and feel insufficiently informed about prognosis and advanced directive However patients report to agree about treatment goals with their physicians in over 90%. Conclusion: These results were fed into the process of consensus building around an ethics policy for advanced care planning and limiting treatment with the participation of representatives from all parties involved in end-of-life decisions: the policy has a special focus and structure on informing patients early about treatment goals and prognosis, while respecting their individual information wishes. Disclosure: No conflict of interest disclosed. P793

Hexamerization of EGFR-directed antibodies induces potent complement-dependent cytotoxicity of tumor cells Tammen A.1, Beurskens F.J.2, Derer S.3, Schwanbeck R.1, Schuurman J.2, Parren P.W.H.I.2, Valerius T.1 Christian-Albrechts-University and University Hospital Schleswig-Holstein, Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, Kiel, Germany, 2Genmab, Utrecht, Netherlands, 3University Hospital Schleswig-Holstein, Medical Department I, Lübeck, Germany 1

Introduction: Monoclonal antibodies are promising therapeutic agents in cancer therapy with the ability to activate the complement (C) system to eliminate tumor cells via the generation of anaphylatoxins, opsonins and membrane-attack complexes. Antibodies directed against the epidermal growth factor receptor (EGFR), a well-validated solid tumor target, lack the capacity to induce complement-dependent cytotoxicity (CDC) as single agents. Recently we showed that optimal C activation and CDC occurs after antigen-bound IgG molecules form hexameric structures through extensive intermolecular Fc-Fc contacts. These hexamers represent an optimal platform for C1q binding and C activation (Diebolder et al, Science 2014, 343, 1260–3). Single point mutations that enhance hexamerization of IgG molecules conditional on antigen binding have been identified. Alternative strategies to increase C activation have focused on enhancing C1q binding via affinity-maturation of the C1q binding site through site-directed mutagenesis or by combining IgG isotypes. The aim of the study was to investigate whether EGFR antibodies optimized for each of these approaches differ in terms of CDC capacity. Methods: EGFR antibodies comprising engineered Fc domains that carry mutations to enhance hexamerization (E345K or E430G), or increase C1q affinity (S239D/ H268F/ S324T/ I332E; DFTE), or that exist of IgG1/IgG3 chimeras (113F) were constructed. CDC activity was assessed by 51Cr release assays in the presence of human serum. Antibody mediated C1q binding or C4b deposition on tumor cells was analyzed by flow cytometry. Apparent C1q binding affinity was determined by C1q specific ELISA. Results: We show that affinity-maturation of the C1q binding site (DFTE & 113F) increased C1q binding in the absence of antigen, whereas mutations that enhance Fc-Fc interactions (E345K & E430G) did not. However, these latter mutations strongly increased C1q avidity and CDC for anti-EGFR molecules bound to cells. Notably, the latter strategy translated in superior CDC activity, which was particularly evident for tumor cells with low EGFR expression levels. Conclusion: Enhancement of hexamer formation by EGFR antibodies conditional on antigen binding is a promising approach to improve their anti-tumor activity. Interestingly, strategies that enhance complement binding independent of antigen engagement by the antibody appear less effective. Disclosure: Annalina Tammen: No conflict of interest disclosed. Thomas Valerius: Expert Testimony: Genmab, Utrecht, The Netherlands.

242

Oncol Res Treat 2015;38(suppl 5):1–270

P794

A novel solid nanoscopic imiquimod suspension enhances tumor rejection induced by transcutaneous immunization Stein P.1, Gogoll K.2, Denny M.2, Weber A.-K.3, Stassen M.3, Langguth P.2, Schild H.3, Radsak M.4 Johannes-Gutenberg University Medical Center, Center for Thrombosis and Haemostasis, Mainz, Germany, 2Johannes-Gutenberg University, Biopharmaceutics and Pharmaceutical Technology, Mainz, Germany, 3JohannesGutenberg University Medical Center, Institute for Immunology, Mainz, Germany, 4Johannes-Gutenberg University Medical Center, Third Department of Medicine, Mainz, Germany 1

Introduction: Transcutaneous immunization (TCI) is a novel vaccination strategy to induce strong therapeutic cytotoxic T-lymphocyte (CTL) responses by directly targeting skin-resident professional antigen-presenting cells (APC) and very promising to overcome current limitations of standard vaccination approaches that are mostly effective in prophylaxis, but not in the treatment of diseases. In this context, we have developed a TCI method based on a synthetic TLR7 agonist imiquimod that mediates partial tumor protection in experimental rodent models. In our present work, we describe a novel optimized formulation of imiquimod in a solid suspension of crystalline imiquimod (IMI-Sol) that induces superior CTL responses and enhanced tumor protection. Methods: We determined imiquimod-penetration into the skin as well as systemic distribution by HPLC in C57BL/6 mice (at 6–12 weeks). For TCI, C57BL/6, Tlr7 (TLR7–/–) or Myd88 (MyD88–/–) gene deficient mice IMI-sol or imiquimod (in the commercially available formulation Aldara®) together with the CTL epitope SIINFEKL (derived from chicken ovalbumin) was applied on two consecutive days on the dorsum of the animals after hair removal by electric clippers. Induction of CTL responses was characterized after 7 days by the analysis of peptide-specific CTLs (ELISpot, tetrameric MHC I complexes) and in vivo cytolytic activity by flow cytometry. Tumor protection was assessed using the B16OVA tumor model. Results: Despite equal imiquimod penetration into the skin, we found superior vaccination capacity of IMI-Sol compared to Aldara® leading to enhanced tumor-protection. Using TLR7–/– or MyD88–/– mice, we confirmed that the induction of CTL responses with IMI-Sol was completely dependent on the TLR/MyD88 pathway. The enhanced immune response is due to more efficiently activated skin-derived DCs upon IMI-Sol treatment found in skin-draining lymph nodes. Conclusions: We developed of a revised imiquimod formulation with superior transcutaneous vaccination efficacy underscoring the importance of pharmaceutical aspects affecting vaccination potency beyond understanding of the underlying basic mechanisms. Combined efforts are necessary to establish TCI as a promising novel next generation vaccination platform that can be used for the treatment of persistent infections and cancer. Disclosure: No conflict of interest disclosed. P795

The Qinghaosu (Artemisinin) derivative Dihydroartemisinin leads to induction of iron catalyzed production of reactive oxygen intermediates in human gastric cancer cells – new treatment strategies from Chinese medicine Löffler C.1, Bosnar S.1, Spielau-Geidies C.1, Einsele H.1, Löffler J.1 Uniklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany

1

Introduction: The prognosis of patients suffering from gastric cancer is dismal. Due to unspecific symptoms, diagnosis occurs late and chances of recovery remain poor. Therefore, development of new treatment strategies is key for improving patient outcomes, especially in palliative settings. The antimalarial sesquiterpen Artemisinin and its derivative Dihydroartemisinin (DHA) have shown first evidence of anti-tumor activity. Underlying mechanisms seem to be quite complex and are not sufficiently understood. The interaction of DHA’s endoperoxide bridge with free iron

Abstracts


CONTENTS AUTHOR INDEX

in Plasmodium falciparum food vacuoles leads to induction of reactive oxygen intermediates (ROI). Due to the fact that tumor cells contain more iron and increased numbers of CD71 transferrin receptor than normal cells, induction of ROI, targeted by ferrous-II-glycine sulphate (Ferro Sanol®) might be a mechanism of anti-cancer toxicity. Therefore, we investigated the in vitro cytotoxic activity of DHA and iron against a human gastric adenocarcinoma cell line (AGS) in combination with potential mechanisms of action. Methods: The cytotoxic effects of DHA were determined by XTT (2,3-Bis(2-Methoxy-4-Nitro-5-Sulfophenyl)-2H-Tetrazolium-5-Carboxanilide) cell viability assay. The absorbance was revealed at 450 nm. To determine the 50% inhibition concentration (IC 50), AGS cells were incubated for 12 hours with DHA in different concentrations (1–100 ug/ml). Oxidative burst was quantified by using dihydrochlorfluorescein diacetate; fluorescence was measured over 2.5 hours at 485/535 nm. To reveal the potential of iron to catalyze DHA mediated ROI production, 10 ug/ml Ferro Sanol® was added. Results: IC 50 was defined to be between 20 and 30 ug/ml DHA (p = 0.019). At concentrations > 90 ug/ml, no AGS survival was observed. ROI release was augmented after stimulation with DHA up to 5.4-fold (p = 0.046). Addition of Ferro Sanol® led to significantly more production of ROI, compared to DHA alone. Time point analysis of ROI production after 100 minutes of DHA/ Ferro Sanol® stimulation showed an increased release (6.6-fold of control; p < 0.01); after 2.5 hours ROI production reached a maximum of 10.4- fold of control (p < 0.01). Conclusion: DHA has shown first evidence of anti-cancer activity towards AGS in vitro. Addition of iron increases this effect. Further experiments have to follow to explore the cytotoxic mechanisms of DHA/iron as a potential add-on therapy in patients with gastric cancer. Disclosure: No conflict of interest disclosed. P796

Willingness to accept randomisation for complementary and alternative medicine (CAM) for breast cancer Heinke H.1, Reimer T.2, Gerber B.2, Markmann S.3, Kirschbaum B.4, Diedrich D.5, Kundt G.5, Freund M.1,6, Junghanß C.1, Lampe H.1,7 Universitätsmedizin Rostock, Klinik für Innere Medizin III, Rostock, Germany, Universitätsmedizin Rostock, Frauenklinik, Rostock, Germany, 3Gynäkologische Praxis, Rostock, Germany, 4Jerusalem Krankenhaus, Mammazentrum, Hamburg, Germany, 5Universitätsmedizin Rostock, Biostatistik und Informatik, Rostock, Germany, 6DGHO, Berlin, Germany, 7Rems-Murr-Klinik Winnenden, Hämatologie, Onkologie und Palliativmedizin, Winnenden, Germany 1 2

Introduction: Treatment of breast cancer is widely standardised, based on large randomised studies. Beside this many patients use complementary treatments. How this influences treatment results is not well known. According to the established treatment concepts randomised studies would be necessary for reliable data. But previous studies have shown that many patients are not willing to accept randomisation. And as most complementary treatments are available over-the-counter, uncontrolled usage may influence treatment results even after randomisation. We investigated how many patients would accept randomisation for complementary treatment. We further evaluated which factors indicate this group of patients. Methods: The survey was carried out at a university hospital and a breast cancer centre. Patients were informed that we conduct a survey to evaluate the acceptance of randomisation and that their data will be processed anonymously. We used a structured questionnaire asking patients to opt for one of three treatment options: 1. Standard treatment. 2. Standard treatment and complementary medicine. 3. Randomisation (“As benefits and risks are not known currently, I would accept random selection.”). We also asked for age, marital status, children, highest qualification, confession, income, other illnesses, malignancies inside the family, knowledge and experience with CAM, from whom they received information on complementary medicine and how satisfied they were with these information. The first 100 incoming questionnaires were used for analyses. The

Abstracts

statistical analyses were performed with SPSS and statistical tests were performed using the chi-square test and the exact test. Results: From 100 patients 76 would opt for standard treatment and complementary medicine, 6 for standard treatment and 18 for randomisation. From analysed factors only previous experience with CAM and satisfaction with consultation on CAM influenced willingness for randomisation. Conclusion: Only 18% of patients would agree to be randomised. Especially patients with previous positive experience with CAM want to decide actively, making it difficult to achieve reliable results on these treatments. It needs reflection that patients unsatisfied with their consultation decide more often for randomisation. It seems unlikely to reach reliable results with randomised studies in the future and we will have to look for alternative ways to cope with these questions (individualised treatment). Disclosure: No conflict of interest disclosed. P797

FungiScopeTM – global emerging fungal infection registry Seidel D.1, Duran Graeff L.2, Wahlers K.2, Vehreschild M.J.G.T.2, Köhler P.2, Müller F.2, Wisplinghoff H.2, Vehreschild J.J.2, Cornely O.A.2 Uniklinik Köln, Infektiologie, Köln, Germany, 2Uniklinik Köln, Cologne, Germany

1

Background: FungiScope™ is a global registry for emerging invasive fungal diseases (IFD) with partners from 60 countries around the globe. The objective is to broaden knowledge on epidemiology of emerging IFD, determine the clinical patterns, describe and improve diagnostic procedures and therapeutic regimens, as well as to facilitate exchange of clinical isolates among the partners. Methods: FungiScope™ uses web-based data capture accessible through www.fungiscope.net. For case enrollment, cultural, histological, antigen or molecular evidence on the occurrence of infection with non-endemic fungi is required. Data collected include demographics, underlying conditions, neutrophil count, immunosuppressive medication, clinical signs, and symptoms, sites of infection, diagnostic tests, pathogen identification, antifungal treatment, surgical procedures, response to treatment, and survival. Results: To date, 429 cases have been captured. Mucorales (n  = 196; 45.7%), Fusarium spp. (n = 65; 15.2%), yeasts (n = 54; 12.6%), and dematiaceae (n = 48; 11.2%) are the most frequently registered pathogens. Chemotherapy (n = 195; 45.5%) and stem cell transplantation for hematological malignancy (n = 102; 23.8%) were the predominant risk factors, followed by intensive care (n = 95; 22.1%), diabetes mellitus (n = 84; 19.6%), and chronic renal disease (n = 35; 8.2%). For 20 cases (4.7%) no risk factor was identified. Sites of infection included lung (n = 214; 49.9%), followed by paranasal sinuses (n = 79; 18.4%), blood stream (n = 77; 17.9%), and deep soft tissue (n = 65; 15.2%). Seventy two cases (16.8%) had disseminated infection, of which 51 cases (70.8%) had lung infection, 31 cases (43.1%) blood stream infection and 24 cases (33.3%) CNS infection. For 212 (52.6%) of all patients, complete or partial response to treatment of IFD was documented. All-cause-mortality and mortality attributable to IFD was 45.2% and 34%, respectively. Conclusion: The clinical relevance of emerging IFD is increasing. In a short time period, a wide variety of cases from around the globe was documented. FungiScope™ turned out to be a vividly expanding network that attained increasing interest throughout the years. Keywords: Rare fungal infections, web-based register, epidemiology Disclosure: Danila Seidel: No conflict of interest disclosed. Oliver A. Cornely: Employment or Leadership Position: Professor of Translational Research.

Oncol Res Treat 2015;38(suppl 5):1–270

243


CONTENTS AUTHOR INDEX

P798

TriReg – study of trichosporonosis Seidel D.1, Wahlers K.1, Vehreschild M.J.G.T.2, Köhler P.1, Liss B.1, Müller F.1, Wisplinghoff H.3, Lass Flörl C.1, Cornely O.A.1 Uniklinik, Köln, Germany, 2UK Köln, Infektiologie, Köln, Germany, 3Universität, Köln, Germany 1

Background: The incidence of invasive fungal infections is increasing in all parts of the world. Less common emerging fungal pathogens account for significant numbers of these invasive infections. Invasive trichosporonosis is one example of an emerging fungal pathogen. To date, the epidemiology is largely unknown and current treatment recommendations cannot be considered evidence based. For this orphan disease, significant numbers of cases for systematic analysis cannot be collected in one center alone. Therefore, “TriReg – A Europewide Study of Trichosporonosis” has been launched as an ECMM (European Confederation of Medical Mycology) Working Group in April 2013. The objective of the study is to overcome the lack of knowledge on invasive trichosporonosis in order to develop evidence-based recommendations for diagnosis and treatment. Methods: TriReg uses web-based data capture via www.ecmm.eu. For case enrolment, cultural, histological, or molecular evidence of invasive trichosporonosis is required. In addition to demographic and clinical data, isolates are collected and formal identification by morphology and molecular tools and susceptibility testing using EUCAST and E-Test are performed. Results: In total 33 cases of invasive trichosporonosis were documented from nine countries. Eighteen patients underwent chemotherapy. Other risk factors were solid organ transplantation (n = 4), stem cell transplantation (n = 4), and abdominal surgery (n = 3). Twelve patients were treated in the intensive care unit. Seventy five percent of the patients experienced disseminated infection documented by positive blood cultures in all cases but one. Outcome was poor with a median survival time of 9 days (range from 1 to 90 days) and an overall mortality rate of 61%. Eighty four percent of the deaths were attributable to the fungal infection. Conclusion: Invasive trichosporonosis is a rare disease with a very poor prognosis. A joint international effort is required to advance the knowledge on this orphan disease, in order to improve patient care. TriReg provides a platform to achieve this goal, and new contributors are invited to participate. Keywords: Trichosporonosis, web-based registry, invasive fungal infection Disclosure: Danila Seidel: No conflict of interest disclosed. Oliver A. Cornely: Employment or Leadership Position: Professor of Translational Research. P799

Patients´ competence in oral cancer therapies Riese C.1, Beylich A.2, Welslau M.3, Benser J.1, Klein A.1, Borges jr. U.1, Zamora P.1, Baumann W.1 Wissenschaftliches Institut der Niedergelassenen Hämatologen und Onkologen (WINHO), Köln, Germany, 2Hämatologisch-onkologische Praxis Altona (HOPA), Hamburg, Germany, 3Hämato-Onkologische Schwerpunktpraxis am Klinikum Aschaffenburg, Aschaffenburg, Germany 1

Introduction: Oral agents in cancer therapy are increasingly prescribed. They are characterized by a considerable potential for side-effects, toxicity and drug interactions. Inadequate use of medication leads to ineffectiveness and in some cases may contribute to an early breakup. Subsequently, patients need a high level of self-management competence. We evaluated whether standardized recurring patient education by oncology nurses influences therapy adherence, self-management ability, and eventually therapeutic success. Methods: The intervention study was conducted in office-based oncology practices in Germany in 2014. Patients starting an oral cancer therapy for the first-time were included. Oncologists in the control group counseled their patients as usual. Oncology nurses in the intervention group were

244

Oncol Res Treat 2015;38(suppl 5):1–270

specially trained to repeatedly provide the patients with information on the clinical picture, side effects, and the proper handling of medication by using the MASCC Oral Agent Teaching Tool (MOATT) in addition to the oncologists counseling. Primary endpoint was the patient´s competence measured by self-efficacy, quality of life and therapy related knowledge. Secondary endpoints were side-effects, health related stress, therapy adherence and breakup rate. Results: In total, 28 office based oncology practices (n = 195 patients, 57% women, 43% men) took part, 17 (n = 136 patients) in the intervention and 11 (n = 59 patients, 50% women, 50% men) in the control group. The mean age of patients in the intervention was 68 years, likewise in the control group. The results reveal a lower therapy interruption rate and a better coping with side-effects in the intervention group. Conclusions: Patients benefit from a standardized patient education program through specially trained oncology nurses. The development of patient centered counseling strategies for oncology nurses may become a corner stone in oral cancer care. Disclosure: No conflict of interest disclosed. P800

What can an oncologist learn from geriatrics? Schroeder M.1,2, Wieschermann U.1, Greiff U.2, Schäfer U.2, Aul C.1 HELIOS St. Johannes Klinik, Klinik für Onkologie u. Hämatologie, Duisburg, Germany, 2HELIOS Marien Klinik, Klinik für Geriatrie u. Innere Medizin, Duisburg, Germany 1

Introduction: Many pts enter our hospital due to decline of general condition and/or suspicion of cancer. The first assessment will have the pts redirected to the appropriate specialists (surgery, gynecology, neurosurgery, gastroenterology). After complete diagnosis pts should be treated according to guidelines by oncologists. Methods: A multidisciplinary team of oncologists and geriatrics decide, following a common visit, a cancer specific geriatric assessment of the SAKK whether to treat the pts in a classic oncologic or geriatric oncolgic department. Results: Between 01/2013 and 12/2014 76 pts (aged 75–92 years, NSCLC:16, breast cancer:17, NHL:18, head&neck:12, ovarian:5, glioblastoma:5, melanoma:3) could be allocated to the geriatric department and could be treated according to Go Go, Slow Go and No Go including geriatric rehabilitation. 58/76 pts saw benefits of increased quality of life and survival time. 56/76 pts relocated to their home with an increased Barthel and Karnofsky index. Thanks to interdisciplinary care discussion a change of the tumorboard decision was made in 31/76 pts. For 85% of these 31 pts less intensive therapy was applied. Further details will be reported. Conclusion: In this field older is not the same as geriatric. Collaboration and cooperation between geriatrics and oncologists means for the older and many geriatric pts remarkable benefit in the treatment and outcome. It also lead to reduced toxicity and side effects. Disclosure: No conflict of interest disclosed. P801

Patients with advanced haematological malignancies in specialized palliative home care (SAPV) Kaiser F.1, von Rudloff L.2, Vehling-Kaiser U.3, Hollburg W.4, Alt-Epping B.2 Universitätsmedizin Göttingen, Klinik für Hämatologie und Onkologie, Göttingen, Germany, 2Universitätsmedizin Göttingen, Klinik für Palliativmedizin, Göttingen, Germany, 3Adiuvantes SAPV, Landshut, Germany, 4PalliativPartner, Hamburg, Germany 1

Introduction: There is sufficient evidence that patients with advanced haematological malignancies in non-curative settings suffer from complex physical symptoms and psychosocial distress, comparable to patients with solid tumor entities. Nevertheless, numerous problems at the interface between haematology and palliative care have been described. This is

Abstracts


CONTENTS AUTHOR INDEX

even more true for palliative home care for patients with haematological malignancies, where virtually no data concerning needs and therapeutic support exist. Methods: We therefore performed a retrospective analysis of all patients with haematological malignancies (ICD 10: C81-C95) being treated by the respective palliative home care team (SAPV), in addition to haematology and general practice. Patient records of three SAPV teams were analyzed (Göttingen, Hamburg-Altona, Landshut); covering all patients from 01/2011 until 10/2014. All SAPV teams were closely neighboured to haematological institutions. Disease entity, demographic data, symptoms, psychosocial distress, length of SAPV care, number of hospital admissions, therapeutic interventions during SAPV and other items were analyzed descriptively. Results: Of 3.955 SAPV patients, only 1.8% (n = 73) suffered from haematological malignancies. Median age was at 76 years, 56% were male and 44% were female. 32% suffered from acute leukaemia, 24% from lymphoma and multiple myeloma, respectively, 16% from chronic lymphocytic leukaemia and 4% from other entities. Main problems were: pain (84%), psychological problems (78%), dyspnea (56%) and deterioration of general condition (52%). 37% developed new symptoms during SAPV care, mainly pain (22%), infections (12%) and weakness (12%). 67% of the patients were not in need of hospitalization. If hospital referral was necessary (33%), there was a wide range of causal, partial combined symptoms, mainly deterioration of general condition (33%) and pain (29%). 70% of the patients died within the first three month after beginning SAPV care and 76% died at home or in a nursing home. Conclusions: Patients suffering from advanced haematological malignancies were statistically underrepresented in SAPV care, and SAPV was predominantly installed at the very last days of life. The spectrum of documented problems resembles to other patient cohorts being treated in SAPV; therefore, the offers and benefits of palliative home care should be incorporated in palliative haematological treatment concepts more vigorously and consequently. Disclosure: No conflict of interest disclosed. P802

Four years of specialized ambulant palliative care (SAPV) in a rural area: cooperation and acceptability from general practitioners´ view Kaiser F.1,2, Sohm M.2, Haas M.2, Gernböck C.2, Illig D.2, Vehling-Kaiser U.2 Universitätsmedizin, Göttingen, Germany, 2Adiuvantes SAPV, Landshut, Germany 1

Introduction: Since 2011, the “Adiuvantes SAPV” team is caring for palliative patients in the counties of Landshut and Dingolfing. In this rural area the cooperation with general practitioners is crucial. Besides caring for patients, building networks for patient-centred care is a substantial part of the SAPV-team´s work. As part of the quality management after four years of SAPV service a survey was performed by questionnaires evaluating the cooperation and acceptance of SAPV among all general practitioners of the respective regions. Methods: From January to March 2015, 194 general practitioners from the counties Landshut and Dingolfing were contacted by questionnaires concerning the issues “training in palliative care”, “cooperation with SAPV”, “saving costs and avoiding hospitalisations”. Next to personal data (gender, age) questions had to be answered by “yes” or “no” or by rating-scales ranging from 1–6. Data were recorded by the coordinating facility of “Adiuvantes SAPV”. Results: We received answered questionnaires from 40 out of 194 contacted general practitioners. Among them 38,2% were females and 61,8% were males. 7.5% of the participants were younger than 40 years, 62.5% were between 40 and 60 years and 30% were older than 60 years. 25% of the colleagues had taken part in a basic training of palliative care, 10% intended to aquire a qualification in palliative care. Furthermore, 10% of the interviewed persons considered an active collaboration in a SAPVteam imaginable, 85% had already cooperated with a SAPV-team. 80,6%

Abstracts

of the participants were absolutely satisfied with patient care provided by the SAPV team, 16,7% were satisfied and 2,7% were not satisfied. 75% stated that hospitalisations were avoided by the inclusion of patients in the SAPV-program whilst 72.5% felt that time and costs were saved for their own practices. Conclusion: The current data show that a training in palliative care amongst general practitioners gains improving importance. The SAPV team has continuously been seeking to include general practitioners in the palliative care. The success of these efforts is reflected by the close collaboration of the SAPV-team with the general practitioners, the high degree of satisfaction of general practitioners with the SAPV-team´s patient care and by the effectiveness concerning time and costs. The current data show the importance of a close collaboration between general practitioners and the SAPV-team. Disclosure: No conflict of interest disclosed.

Posterdiskussion Sarkom P803

An updated overall survival analysis with correction for protocol-planned crossover of the international, phase III, randomized, placebo-controlled trial of regorafenib in advanced gastrointestinal stromal tumors after failure of imatinib and sunitinib (GRID) Grünwald V.1, Demetri G.D.2, Reichardt P.3, Kang Y.-K.4, Blay J.-Y.5, Joensuu H.6, Schäfer K.7, Kuss I.7, Kappeler C.7, Casali P.G.8 Medizinische Hochschule Hannover, Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Hannover, Germany, 2Ludwig Center at Dana-Farber Cancer Institute and Harvard medical School, Boston, United States, 3Helios-Klinikum, Berlin, Germany, 4University of Ulsan College of Medicine, Seoul, Korea, Republic of, 5Léon Bérard Centre and Claude Bernard University, Lyon, France, 6Helsinki University Central Hospital, Helsinki, Finland, 7Bayer Pharma AG, Berlin, Germany, 8National Cancer Center Hospital EastInstitute, Milan, Italy 1

Introduction: The GRID study showed that regorafenib improves progression-free survival compared with placebo in patients with advanced GIST after failure of at least imatinib and sunitinib (HR 0.27; 1-sided p_0.0001; Demetri 2013). At the time of the primary analysis, no significant difference in the secondary endpoint of overall survival (OS) was observed (HR 0.77; p_0.199), but this result may have been confounded by the high rate of crossover to regorafenib (85%) of placebo patients at progression. We conducted exploratory analyses of updated OS data to assess the effect of correcting for this protocol-planned crossover. Methods: The data cut-off for this updated OS analysis was 31 January 2014 (2 years after the primary analysis). OS was corrected using two randomization-based methods: rank preserving structural failure time (RPSFT) and iterative parameter estimation (IPE); both methods are considered as best choice among all correction analytics. Hazard ratios and 95% CI were derived using the Cox model. Results: A total of 139 deaths had occurred at the time of data cut-off: 91 events (68.4% of patients) in the regorafenib group and 48 (72.7%) in the placebo group. A total of 22 patients remained on regorafenib treatment (median duration 2.1 years, range 0.9–2.4). The updated hazard ratio for OS favored regorafenib (0.85, 95% CI: 0.60–1.21; p_0.18). Median OS was estimated as 17.4 months in both groups, with crossover from placebo. The corrected HRs for OS are less than the uncorrected HR (Table). Conclusion: The updated analysis of OS in the GRID trial is consistent with the primary analysis. An exploratory analysis correcting for the impact of cross-over on OS suggests a survival benefit for regorafenib in GIST. Clinical trial information: NCT01271712.

Oncol Res Treat 2015;38(suppl 5):1–270

245


CONTENTS AUTHOR INDEX

P805

Tab. 1. Overall Survival Analyses

OS crossover correction method

Hazard Ratio

95% CI

ITT, uncorrected analysis

0.85

0.60–1.21

RPSFT

0.39

0.26–0.58

IPE

0.51

0.35–0.73

Disclosure: Viktor Grünwald: Advisory Role: Bayer, Pfizer. Paolo Casali: No conflict of interest disclosed. P804

Solitary fibrous tumors / haemangiopericytoma: Analysis of prognostic factors of 28 patients from a high volume sarcoma center Virchow I.1,2, Podleska L.1,3, Pöttgen C.1,4, Reis A.C.1,5, Steinau H.-U.1,3, Taeger G.1,3, Farzaliyev F.1,3, Schuler M.2,6, Bauer S.1,2,6 Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany, 2Department of Medical Oncology, Essen, Germany, 3Department of Surgery, Essen, Germany, 4Department of Radiotherapy, Essen, Germany, 5Institute of Pathology and Neuropathology, Medical Faculty, University of Duisburg-Essen, Germany, Essen, Germany, 6 German Cancer Consortium (DKTK), Heidelberg, Germany 1

Introduction: Haemangiopericytomas have recently been added to the group of (extrapleural) solitary fibrous tumours (SFT), which belong to the fibroblastic/myofibroblastic tumor category. SFT show heterogenous clinical features that include both indolent and aggressive behavior as well as unpredictable response to classical chemotherapies. We sought to analyze the clinical characteristics of a larger series of SFT treated at a single institution. Methods: The institutional database (n = 2850) was queried for the term SFT which resulted in 28 cases (1%). Data on clinico-pathologic parameters, disease recurrence, location of metastases, systemic therapy and survival were collected from a retrospective chart review. Overall survival curves were calculated using Kaplan-Meier analyses. Results: The cohort consisted of 68% male patients with a median age at diagnosis of 53 years (21–77 years). Median follow up was 69mo. Location of primaries were trunk (46%), head (29%) and extremities (25%) with a median tumor size of 7.5cm. Local recurrence occurred in 50% notably not at extremity site. 54% of pts developed metastases of which 87% were metachronous. 47% of patients had both pulmonary and extrapulmonary disease and 33% showed extrapulmonary sites only. The majority of patients received both classical chemotherapeutic therapies and targeted treatments. Median OS was not reached for the whole population. Median time to local relapse or metastatic disease was 7.7 years. Conclusions: In our series, we observed a high rate of metastatic disease which contrasts to the term “solitary” fibrous tumors but a prolonged overall survival despite metastatic spread. SFTs more commonly occur at non-extremity sites and show a high rate of extrapulmonary metastases. Local recurrence and metastases may occur after long-term disease-free period which suggests a prolonged FU for these patients. Aggressive local therapies should be considered at all primary tumor sites. Disclosure: No conflict of interest disclosed.

A pooled shRNA lentiviral screening model to identify EWS-FLI1 specific therapeutic targets in Ewing sarcoma Schaefer C.1, Berning P.1, Mallela N.2, Seggewiß J.3, Schleithoff C.1, Korsching E.2, Dirksen U.1, Potratz J.1 Universitätsklinikum Münster/Klinik für Kinder- und Jugendmedizin, Münster, Germany, 2Westfälische Wilhelms-Universität Münster/Institut für Bioinformatik, Münster, Germany, 3Universitätsklinikum Münster/Institut für Humangenetik, Münster, Germany 1

Introduction: Ewing sarcomas are high risk sarcomas that require novel treatment approaches to improve survival. Ewing sarcoma is defined by characteristic genetic alterations that lead to oncogenic transcription factors, EWS-FLI1 in most cases. As direct targeting of EWS-FLI1 remains problematic, we aim to target signaling pathways that cooperate in cellular EWS-FLI1 tolerance as an alternative approach to tumor-cell specific therapies. To identify such pathways we established a pooled lentivral shRNA screening model. Methods: To identify targets specific for the presence of EWS-FLI1 we utilized an A673 cell line model stably bearing shRNA directed at EWS-FLI1 (off) or non-expressed ERG control (on). The pooled screening approach is based on enrichment and depletion of shRNA from a cell pool, with depleted shRNA representing indispensable proteins and pathways as potential therapeutic targets. shRNA frequency and identity are identified by Next-Generation sequencing. A Decode Pooled GIPZ lentiviral shRNA library encompassing 4.675 shRNA directed at 709 human kinases was screened. Results: Transduction conditions of A673 cells were optimized for maximum efficacy using self-generated and purchased lentiviral particles. As pool deconvolution requires a calculated transduction of one shRNA per cell, titration experiments were performed to adjust the multiplicity of infection to 0.3. Effective single shRNA-copy protein knockdown was confirmed. shRNA were transduced at a 100-fold representation which was maintained throughout the screening procedure. Cell populations were selected for survival under standard cell culture conditions. Genomic DNA was isolated from cell populations and viral input control and integrated shRNA sequences were amplified. Barcoding and screening was adapted to the IonProton platform. Preliminary alignments confirmed correlation of technical replicates and deletion of a set of shRNA from experimental samples, representing screening hits. Further results will be presented. Conclusion: The method established provides a versatile functional tool to screen for therapeutic targets. In the setup screened here it will identify tumor-cell specific targets synthetic lethal to the presence of EWS-FLI1 in Ewing sarcoma. Acknowledgements: The A673 cell line model was provided by S. Lessnick (Huntsman Cancer Institute, University of Utah), financial support was by BMBF (FKZ 01GM0869), ERA-Net-TRANSCAN (01KT1310) and EEC (602856–2). Disclosure: No conflict of interest disclosed. P806

Intratumoral immune cell infiltration in patients with highgrade soft-tissue sarcoma is associated with tumor grading Bücklein V.1,2, Kampmann E.1, Noessner E.3, Issels R.1, Lindner L.1, Subklewe M.1,2, Knösel T.4 Klinikum der Universität München, Medizinische Klinik und Poliklinik III, München, Germany, 2Helmholtz Zentrum München, KKG Immuntherapie, München, Germany, 3Helmholtz Zentrum München, Institut für Molekulare Immunologie, München, Germany, 4Institut für Pathologie, LudwigsMaximilians-Universität, München, Germany 1

Introduction: In recent years, the immune system has been shown to play an important role in the development and progression of cancer. For many kinds of cancer, infiltration with immunocompetent cells has prognostic significance. Soft-tissue sarcomas (STS) are a heterogeneous group

246

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

of rare mesenchymal malignancies. Known prognostic factors for STS include size, localisation and grading. The aim of the study was to evaluate the prognostic importance of tumor-infiltrating lymphocytes and different T-cell subsets for overall and progression free survival, together with their association with the grading of the tumors. Patients and methods: Tissue microarrays (TMA) of primary tumor samples of 210 STS patients were assessed for lymphocyte infiltration, counted in standard HE stained TMAs. Additionally, immunohistochemistry stainings for CD3 (a pan-T-cell marker), FOXP3 (a marker for regulatory T cells) and PD-1 (a marker for T-cell exhaustion) were evaluated. For 68 patients, tumor samples were available before and after neoadjuvant chemotherapy. Cells were counted semiquantitatively, and results were analyzed for associations with overall/progression free survival and grading. Additionally, differences in cell counts before and after neoadjuvant treatment were assessed. Results: Tumor infiltration with lymphocytes, CD3-positive and FOXP3-positive cells was significantly correlated with tumor grading. Median progression free survival was significantly shorter for patients with >2 lymphocytes/TMA (median 58 vs. 129 months, p = 0,014). For CD3, FOXP3 and PD-1-positive cells, differences in tumor infiltration did not result in significantly different survival. Neoadjuvant therapy resulted in a significant increase in lymphocyte numbers within the tumor. CD3-positive cells and PD-1-positive cells did not change, whereas counts for FOXP3-positive cells were reduced after treatment. Interestingly, in contrast to the pre-treatment counts, high intratumoral lymphocyte counts after treatment were not associated with shorter survival. Conclusions: Intratumoral lymphocyte infiltration, assessed before the start of neoadjuvant treatment, in STS patients is a negative prognostic factor. Although neoadjuvant treatment further increases the lymphocyte infiltration and leads to significant changes in the distribution of T cell subsets within the tumor, high lymphocyte counts after neoadjuvant treatment are not associated with shorter survival any more. Disclosure: No conflict of interest disclosed. P807

Gene expression analyses of receptor tyrosine kinases identify ROR1 as a potential therapeutic target in metastatic Ewing sarcoma Berning P.1, Tillmanns A.1, Korsching E.2, Lechtape B.1, Schaefer C.1, Schleithoff C.1, Schäfer K.-L.3, Dirksen U.1, Potratz J.1 Universitätsklinikum Münster, Klinik für Kinder- und Jugendmedizin, Münster, Germany, 2Westfälische Wilhelms-Universität Münster, Institut für Bioinformatik, Münster, Germany, 3Universitätsklinikum Düsseldorf, Institut für Pathologie, Düsseldorf, Germany 1

Introduction: Despite most intensive treatments, prognosis for patients with metastatic Ewing sarcoma remains poor and novel strategies are needed. In the development of agents directed at molecular cancer hallmarks, receptor tyrosine kinases (RTK) have provided several targets. IGF1R-directed approaches confirm this in principle for Ewing sarcoma. But, emerging evidence on redundancy and cross-signaling demonstrates RTK signaling as a dynamic network. In one approach towards a better understanding of the RTK network in Ewing sarcoma, we performed a comprehensive gene expression analysis. Methods: In a semi-automated setup, high-throughput probe-based RT-PCR was performed on 55 RTK in 8 Ewing sarcoma cell lines. Subsequently, 45 expressed RTK were analyzed in 21 samples of untreated Ewing sarcoma primary tumors. 6 of these were from metastatic and multifocal disease, 15 from localized disease. Mesenchymal stem cells (MSC) served as control. Results: Expression of all RTK families and multiple family members confirms a potential for redundancy. Highest over-expression compared to MSC and most frequent high-level expression across all RTK was ob-

Abstracts

served for VEGFR3 and TIE1 receptors, highlighting angiogenic signaling axes. High-level expression of these RTK and of EPHA7, ERBB3, and RYK was also suggestive of poor prognosis in localized tumors. A global high-level RTK expression was linked to metastatic disease and 9 individual RTK showed significant over-expression in metastatic Ewing sarcomas. Among these was ROR1, an emerging target in leukemia and carcinomas, where it promotes metastasis. We confirmed ROR1 cell surface expression in Ewing sarcoma cell lines. A so-considered active ROR1 isoform was phosphorylated. In contrast to a carcinoma model this was independent of concomitant activation of the MET receptor. In contrast Wnt5a, a suggested ROR1 ligand, promoted cellular migration. ROR1 silencing reduced migration, confirming a role in a metastatic cellular feature. Conclusion: This comprehensive receptor tyrosine kinase gene expression analysis provides a basis for further study of the therapy-relevant RTK network in Ewing sarcoma. Also, we identify ROR1 as a receptor tyrosine kinase with metastasis-associated cellular function. Presuming additional studies, Ewing sarcoma patients may profit from an ongoing development of ROR1 targeted therapeutics in more frequent cancers. Financial supported by ERA-Net-TRANSCAN [01KT1310] and EEC [602856–2]. Disclosure: No conflict of interest disclosed. P808

TNM-classification for clear cell sarcomas – is it useful? Mertins S.1, Virchow I.1, Farzaliyev F.2, Geismar D.3, Podleska L.2, Pöttgen C.4, Reis A.-C.5, Steinau H.-U.2, Stuschke M.4, Taeger G.2, Treckmann J.6, Schuler M.1, Bauer S.1 Universitätsklinikum Essen, Innere Klinik (Tumorforschung), Essen, Germany, Universitätsklinikum Essen, Klinik für Orthopädie und Unfallchirurgie, Essen, Germany, 3Universitätsklinikum Essen, Westdeutsches Protonenzentrum, Essen, Germany, 4Universitätsklinikum Essen, Klinik für Strahlentherapie, Essen, Germany, 5Universitätsklinikum Essen, Institut für Pathologie, Essen, Germany, 6Universitätsklinikum Essen, Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Essen, Germany 1 2

Introduction: Clear cell sarcomas are one of the most infrequent sarcomas. They exhibit unique clinical characteristics when compared to other sarcomas subtypes such as frequent regional lymph node involvement and frequent metastatic spread despite small size and overall poor prognosis. We sought to analyze clinical characteristics and outcome of a larger cohort of clear cell sarcomas in a high volume sarcoma center. Methods: The institutional sarcoma database (n = 2850) was queried for clear cell sarcomas and clinical data was collected by retrospective chart review. Covariates were tumor size, TNM-status, age of diagnosis, time to metastasis, response and duration of systemic treatments and overall survival. Kaplan-Meier-curves were calculated using SPSS 19.0. Results: We identified 31 patients with clear cell sarcoma amounting to only 1% of all sarcomas. The cohort consisted of 15 male and 16 female patients. The median age at diagnosis was 38.8 years. 90% of tumors were located on the extremities with 60% occurring in a distal location. The average tumor size was 3cm with 60% of patients having T1 tumors (<5 cm). Notably, no difference in outcome was found in comparison with patients who had T2 tumors. 57% of all patients developed metastatic disease. The median time to metastatic disease was 6 years after diagnosis in localized patients; synchronous metastases were found in 16% of all cases. Median overall survival was 7.1 years with a 5 years overall survival rate of 55%. Classical chemotherapies (such as doxorubicin and ifosfamide) showed a clinical benefit in only 10% of the patients that were treated. Conclusions: Clear cell sarcomas frequently arise in the distal extremity, are small in size and diagnosed in mostly young patients. The TNM-classification most likely underestimates the risk of metastatic spread as patients develop metastases even after resection of small T1-tumors. Our data suggest the development of a clear cell sarcoma-specific staging system that should explore additional molecular predictive markers. Adjuvant treatment studies should incorporate tumors of every size. Clinical

Oncol Res Treat 2015;38(suppl 5):1–270

247


CONTENTS AUTHOR INDEX

trials should be considered standard of care as classical chemotherapy shows limited therapeutic benefit. Disclosure: No conflict of interest disclosed. P809

Patients with soft tissue sarcoma comprise a higher frequency of comorbidities than cancer-free individuals – a secondary data analysis Trautmann F.1, Singer S.2, Schmitt J.1 Universitätsklinikum Carl Gustav Carus Dresden, Zentrum für evidenzbasierte Gesundheitsversorgung, Dresden, Germany, 2Universitätsklinikum Mainz, Institut für medizinische Biometrie, Epidemiologie und Informatik (IMBEI), Mainz, Germany 1

Background: Soft tissue sarcoma (STS) comprise a heterogeneous group of solid malignant tumors located in the soft and fat tissues. Comorbidities are important prognostic factors for survival and adversely impact quality of life. We examined the complex relationship between STS and comorbidities over time in a large population-based sample. Methods: The study is based on routine data of the German statutory health insurance AOK PLUS covering longitudinal data of overall 2,615,865 insured persons from 2005 till 2012. Case identification of STS and comorbid diseases was based on respective ICD-10-GM codes and diagnostic modifiers. Uni- and multivariate regression models were used to obtain risk estimates for chronic somatic and mental comorbidities in STS patients compared to a cancer-free control group. Results: At diagnosis, patients with STS were more likely to be affected with prevalent bronchial asthma (OR = 1.6, 95% CI: 1.2–2.1), ≥ one cardiovascular risk factor (OR = 1.7, 95% CI: 1.4–2.1), back pain (OR = 1.3, 95% CI: 1.1–1.5), depression (OR = 1.7, 95% CI: 1.4–2.1), anxiety disorder (OR = 1.8, 95% CI: 1.3–2.3), and adjustment disorder (OR = 2.0, 95% CI: 1.4–2.8) than cancer-free controls. During the course of STS disease, they were at higher risk to develop incident depression (RR = 1.7, 95% CI: 1.2–2.3), anxiety disorder (RR = 1.9, 95% CI: 1.3–2.9), and adjustment disorder (RR = 2.8, 95% CI: 1.9–3.9) than cancer-free individuals. Conclusion: Our results suggest comorbidities to be more often existent in STS patients compared to cancer-free controls of the same age and sex. Comorbidities need to be considered in clinical decision-making regarding the treatment of STS patients. Psycho-oncological treatment should be incorporated into medical care of patients with sarcoma. Disclosure: No conflict of interest disclosed. P810

chemotherapy with vincristine, ifosfamide plus alternating doxorubicin and dactinomycin (VAIA3) was applied. After three cycles, morphologic and metabolic remission was confirmed by FDG-PET/CT. Therefore secondary surgical resection is planned after six cycles of chemotherapy. The results of whole-exome and transcriptome sequencing to characterize the tumor on the molecular level and to explore potentially therapeutic targets are still pending. This is – to our knowledge – one of the few reports world-wide on resectability following neoadjuvant chemotherapy in AFH, indicating a potential role of chemotherapy in this disease. Disclosure: No conflict of interest disclosed. P811

An extragastrointestinal stromal tumor in the mesentery Adamova Z.1, Sankot J.1, Bár T.1, Šindler P.1 Nemocnice Vsetin a.s., Vsetin, Czech Republic

1

Introduction: Gastrointestinal stromal tumors (GISTs) represent the majority of primary nonepithelial neoplasms of the digestive tract. Extragastrointestinal stromal tumors (EGISTs) are neoplasms with overlapping immunohistological features, occurring in the abdomen outside the gastrointestinal tract with no connection to the gastric or intestinal wall, in sites including the omentum, mesentery, pleura and retroperitoneum. Methods: We report a case of a large EGIST in a 68-year-old male. We describe its histopathological and clinical features and the course of our treatment. Results – case presentation: Our patient underwent right hemicolectomy for adenocarcinoma in 2008. A small mass arising from the wall of ileum was incidentally found during this operation, histological examination revealed GIST of benign character. Five years after the first surgery he was operated again, this time because of large tumor in mesentery. We resected it en bloc with the adjacent jejunum. According to the size, mitotic activity, cellularity, necrotic situation and immunohistochemical data, the EGIST belonged to a high-risk group. Abdominal computed tomography performed again after surgery showed four small liver metastases. The patient received targeted therapy (imatinib 400 mg, daily). No recurrence or new metastasis has been identified during two years of follow up observations. Conclusions: EGIST can grow slowly in the abdomen for a long time without clinical appearance. In most cases a preoperative diagnosis is not possible, and the patient undergoes a surgical operation for the diagnosis of “abdominal mass”. During the intervention it is important to achieve a complete removal of the mass.

Multimodal therapy of a recurrent metastatic angiomatoid fibrous histiocytoma: A case report

Disclosure: No conflict of interest disclosed.

Richter S.1, Radke J.1, Fröhling S.2, Platzek I.3, Beuthien-Baumann B.4, Weitz J.5, Ehninger G.1, Folprecht G.1

Posterdiskussion

University Hospital, Dresden University of Technology, Medical Department I, Dresden, Germany, 2National Center for Tumor Diseases and German Cancer Research Center, Department of Translational Oncology, Heidelberg, Germany, 3 Dresden University of Technology, Department of Radiology, Dresden, Germany, 4Dresden University of Technology, Department of Nuclear Medicine, Dresden, Germany, 5University Hospital, Dresden University of Technology, Department for Visceral, Thoracic and Vascular Surgery, Dresden, Germany 1

Angiomatoid malignant fibrous histiocytoma (AFH) is a rare soft-tissue tumor of intermediate malignancy that is most commonly found in children, adolescents and young adults, that is distinct from MFH. Tumors are often located in the extremities, trunk and head and neck usually treated by wide excision. Metastatic behavior is very uncommon and therefore, there is only very limited data on the role of radiotherapy or chemotherapy for unresectable or metastatic disease. Here, we present the case of an 18-year-old male patient suffering from recurrent AFH with multiple intraabdominal and retroperitoneal metastases six months after wide excision of an intraabdominal AFH. Neoadjuvant

248

Oncol Res Treat 2015;38(suppl 5):1–270

Sonstige Themen II P812

Influence of the novel proteasome inhibitor carfilzomib on human monocyte-derived dendritic cells Dahlfrancis M.1, Dörfel D.1,2, Stickel J.S.1, Walz S.3, Grünebach F.1, Rittig S.M.1 Uniklinik Tübingen, Abteilung für Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Tübingen, Germany, 3Universität Tübingen, Interfakultäres Institut für Zellbiologie, Bereich Immunologie, Tübingen, Germany 1

Introduction: The introduction of immune-modulatory drugs and proteasome inhibitors have significantly improved prognosis of multiple myeloma (MM). However, with most patients developing relapse elimination of residual malignant cells is crucial in achieving cure. T-cell based im-

Abstracts


CONTENTS AUTHOR INDEX

munotherapy alone or in combination with novel agents may represent a well-tolerated treatment option, particularly since myeloma cells have shown to be valid targets providing a panel of naturally presented myeloma-associated antigens. Dendritic cells as the most potent antigen-presenting cells play a central role in T-cell based immunotherapy. In order to evaluate the potential of combinatory approaches we analyzed the influence of carfilzomib, a novel proteasome inhibitor approved for treatment of MM on human monocyte-derived dendritic cells (moDC). Methods: MoDC were generated from peripheral blood mononuclear cells by plastic adherence and exposure to GM-CSF (100 ng/ml) and IL-4 (20 ng/ml). Clinically relevant concentrations of carfilzomib (3 nM, 30 nM, 100 nM) were added to the culture on day 6 for 1 h. Bortezomib (1 ng/ml for 24 h) served as positive control. Cells were harvested for immunophenotyping using flow cytometry and for migration assays on day 6 (immature moDC) or after exposure to LPS (100 ng/ml) on day 7 (mature moDC). Results: Immunophenotypic analyses revealed no significant difference in expression of HLA-DR and DC-SIGN and the co-stimulatory molecules CD80 and CD86. Moreover, neither the maturation marker CD83 nor the monocyte marker CD14 showed differences under the influence of carfilzomib as compared to the negative control, indicating no significant influence on phenotype and maturation of human moDC (n = 6). In order to evaluate possible influence of carfilzomib on DC function migration assays were performed. In line with FACS analyses no significant changes in migratory capacity were observed when carfilzomib was added to the DC culture (n = 5). Conclusion: Our results indicate that carfilzomib does not influence DC phenotype or prevent maturation in comparison to bortezomib. Furthermore, migratory capacity is not significantly impaired indicating that combinatory approaches of DC-based immunotherapy and treatment with the proteasome inhibitor carfilzomib should be feasible. Disclosure: No conflict of interest disclosed. P813

The E3 ubiquitin ligase Cbl-b limits Th9 differentiation Cornez I.1, Parampalli Yajnanaraya S.1, Schmidt S.2, Garbi N.3, Brossart P.1, Wolf D.1 University Clinic Bonn, Department of Internal Medicine III, Oncology, Hematology and Rheumatology, Bonn, Germany, 2Innsbruck Medical University, Internal Medicine V, Innsbruck, Austria, 3University Clinic Bonn, Institute of Molecular Medicine and Experimental Immunology (IMMEI), Bonn, Germany 1

Introduction: Th9 cells are critical mediators of anti-cancer immunity and allergy. The E3 ubiquitin ligase Cbl-b modulates T cell activation via regulation of the T cell receptor (TCR) activation threshold as well as by controlling TGFβ-sensitivity, which is critically involved in Th9 differentiation. In this study, we evaluate the role of Cbl-b in TGFβ-dependent Th9 differentiation. Methods: Th9 cells were generated from WT and cblb-deficient naïve CD4+ T cells. Differentiation was determined by quantification of cytokines, mainly IL-9, and the two transcription factors required for Th9 differentiation, namely IRF4 and PU.1. Microarray assay revealed gene candidates that were further validated by mRNA and protein expression analysis. The functional role of Cbl-b was tested in a Th9-mediated murine lung allergy model, in which mice were challenged by intratracheal injections of house dust mite extracts. Results: cblb-deficient naïve T cells are more efficiently differentiated into Th9 cells, express PU.1 more rapidly and intensively, while retaining similar expression levels of IRF4 than WT Th9 cells. Microarray analysis revealed that RUNX1, a transcriptional repressor of PU.1, is more rapidly down-regulated in cblb-deficient Th9 cells than in WT Th9 cells. When knocking down RUNX1 by siRNA in naïve CD4+ T cells and subsequently differentiating them into Th9 cells, RUNX1-depleted Th9 cells present a higher IL-9 expression at the mRNA and protein levels than control Th9 cells. In a mouse allergy model, cblb-deficient mice have a higher lung inflammation as mirrored by increased eosinophils in the BAL and in the

Abstracts

lungs as well as increased IgE production, which is paralleled by an increased IL-9 expression level in the lungs of allergic cblb-deficient mice. Conclusions: Cbl-b critically limits Th9 differentiation and may thus be a potential target to modify Th9 cell generation in allergy or cancer. Disclosure: Isabelle Cornez: No conflict of interest disclosed. Dominik Wolf: Financing of Scientific Research: Novartis, BMS, Pfizer, Amgen, AOP; Expert Testimony: Novartis, AOP. P814

RAC1 selectively engages the β isoform of PI3K in PTEN loss Fritsch R.1, Fritsch K.1, Sommer S.1, DeKrijger I.2, Downward J.2 Uniklinik Freiburg, Innere Medizin I, Freiburg, Germany, 2Cancer Research UK London Research Institute, London, United Kingdom 1

The PIP3 phosphatase PTEN is very frequently inactivated in human cancer by somatic mutation, allelic deletion or epigenetic downregulation. Loss of PTEN leads to the constitutive hyperactivation of the class I phosphoinositide 3-kinase (PI3K) pathway, which in turn promotes many aspects of the malignant phenotype, including uncontrolled growth, apoptosis resistance, invasiveness and metastatic spread. Genetic studies and cell line based data point to a predominant role of the ubiquitous p110β isoform of PI3K in PTEN-deficient tumours (1–3), for which isoform-selective p110β inhibitors are now in clinical trials (NCT01458067, NCT01884285, NCT01673737). The molecular mechanisms underlying the particular importance of p110β in PTEN loss have remained largely obscure. We previously reported that p110β is uniquely regulated through direct binding of the RHO family GTPases RAC1 and CDC42 to its RAS-binding domain (RBD). We here show that p110β is co-activated by RAC1 in PTEN loss. Genetic disruption of the RAC1-p110β signalling axis blocks PTEN loss-induced transformation and invasion in vitro and impairs PTEN loss-driven tumourigenesis in murine prostates. Mechanistically, PTEN negative cells show enhanced steady-state RAC activity and PTEN null prostates exhibit strong RAC-p110β interaction in vivo. Finally, pharmacological inhibition of RAC inhibits PI3K activity selectively in PTEN null human cancer cell lines. Taken together, our findings establish RAC1 as critical molecular link between PTEN loss and PI3Kp110β activity. Disclosure: No conflict of interest disclosed. P815

C/EBPα -induced proinflammatory cytokines regulate early dendritic cell development Rosenberger A.1, Geiger O.1, Zebisch A.1, Sill H.1, Wölfler A.1 Medizinische Universität Graz, Klin. Abteilung für Hämatologie, Graz, Austria

1

Introduction: The transcription factor C/EBPα is crucially involved in myelopoiesis. However, its role in dendritic cell (DC) development is less clear. The aim of this project was therefore to elucidate a role of C/EBPα in DC development. Methods: Bone marrow (BM) cells were obtained either from Mx1Cre CebpaF/F mice, which have a BM specific knock-out (ko) of C/EBPα and consecutive loss of all mature myeloid cells after pIpC treatment, or CebpaF/F control mice. DC formation was modelled in vitro by incubation of lineage negative BM progenitors with FLT3L for 8 days and analysed by flow cytometry using markers of mature DCs, apoptosis was detected by annexin V/propidium iodide staining. Gene expression profiling was performed using Affymetrix GeneChip Mouse Gene 1.0 ST arrays. Cytokine and chemokine production in vitro was measured using Illumina multiplex assays. Results: While BM progenitors isolated from wt mice proliferated and differentiated into mature DCs, Cebpa ko BM progenitors displayed increased apoptosis and DC formation was almost absent. Gene expression analysis of FLT3L-stimulated BM progenitors showed not only C/EBPα-dependent upregulation of DC-specific genes, but also upregulation of TNFα, IL-1β, several chemokines, such as MIP-1α and MIP-2, as well as

Oncol Res Treat 2015;38(suppl 5):1–270

249


CONTENTS AUTHOR INDEX

NF-κB-dependent genes including the anti-apoptotic factor Bcl2A1. Furthermore, production of TNFα, IL-1β, MIP-1α and MIP-2 was detected in the supernatant of BM cells from wt mice but not in Cebpa ko BM progenitors. Interestingly, while addition of IL-1β to FLT3L increased survival of Cebpa ko BM progenitors, addition of TNFα not only restored survival but also mature DC formation. In contrast, addition of MIP-1α and MIP-2 did not restore survival or DC maturation in Cebpa ko cells. Conclusions: We identified a critical role of C/EBPα in regulating production of proinflammatory cytokines, such as TNFα and IL-1β in BM progenitors. Via this mechanism C/EBPα may affect NF-κB-dependent survival and maturation during DC development. Disclosure: No conflict of interest disclosed. P816

5´-deoxy-5´-methylthioadenosine suppresses human T cell function and represents a novel mechanism of tumor immune escape Strobl C.1, Henrich F.1, Singer K.2, Gronwald W.3, Bosserhoff A.4, Kreutz M.2, Poller K.1, Kremer A.1, Mackensen A.1, Aigner M.1 Universitätsklinikum Erlangen; Hämatologie und Onkologie, Erlangen, Germany, 2Universitätsklinikum Regensburg; Hämatologie und Onkologie, Regensburg, Germany, 3Universität Regensburg, Institut für Funktionale Genomik, Regensburg, Germany, 4Friedrich-Alexander Universität ErlangenNürnberg, Institut für Biochemie, Erlangen, Germany 1

Introduction: Creation of an immunosuppressive microenvironment is one mechanism of tumor immune escape. Amongst others, tumors can take advantage of their metabolic alterations to inhibit immune responses. In recent years, methylthioadenosine phosphorylase (MTAP) deficiency gained more and more interest as putative immuno-inhibitory metabolic dysregulation. Loss of MTAP is found in various tumor entities and leads to accumulation of 5’-deoxy-5’-methylthioadenosine (MTA). Lack of MTAP is associated with an inferior response towards adjuvant interferon-α therapy and a higher risk for metastatic disease in malignant melanoma. Here we examined the role of tumor-secreted MTA in suppression of anti-tumor T cell responses. Methods: The effect of MTA was investigated in human polyclonal or tumor antigen-specific T cells. As a read-out we evaluated proliferation, viability, activation, differentiation, clonal expansion and effector function of the T cells. In addition, co-culture experiments of T cells with a MTA-secreting tumor cell line were performed. Finally, we used retroviral transduction to generate MTAP-overexpressing T cells as a putative strategy to overcome MTA-mediated inhibitory effects. Results: MTA strongly reduced proliferation, activation, and viability of stimulated T cells in a dose-dependent manner. Next, induction and expansion of antigen-specific T cells in an autologous co-culture system as well as differentiation and effector functions of antigen-specific T cells were significantly reduced upon MTA challenge. Moreover, MTA-mediated suppression could be confirmed using an MTA-secreting tumor cell line, which inhibited T cell proliferation in mixed lymphocyte/tumor cell cultures. Mechanistically, we found MTA to interfere with Akt signaling and protein methylation, both critical factors for proper T cell function. Of interest, we successfully generated stable MTAP-overexpressing T cells. Preliminary experiments revealed reduced cell death of MTAP-overexpressing T cells upon co-culture with MTA compared to mock control suggesting a promising approach to reconstitute inhibition by MTA. Conclusions: Our data emphasizes the importance of tumor metabolites such as MTA in the tumor milieu and provides a novel strategy of tumor immune escape. Further characterization of the molecular mechanisms of MTA-induced immunosuppression will help to develop more effective immune-based therapy against MTAP-deficient tumors. Disclosure: No conflict of interest disclosed.

250

Oncol Res Treat 2015;38(suppl 5):1–270

P817

Phosphatase and tensin homolog (PTEN) promotes hypoxiainducible factor-2α-dependent amphiregulin production in PTEN-deficient glioblastoma cells U87MG Sun W.1, Jelkmann W.1, Depping R.1 Universität zu Lübeck, Institut für Physiologie, Lübeck, Germany

1

Introduction: Hypoxia-inducible factor-2α (HIF-2α) is the oxygen-labile α-subunit of hypoxia-inducible factor (HIF), a transcription factor which mediates the cellular transcriptional responses to oxygen deprivation (hypoxia). The stability of HIF-2α is dependent on oxygen concentration. Besides regulating the cellular adaptation to hypoxia, HIF-2α has also been shown to promote the growth of glioblastoma cells by maintaining the expression of the auto-/paracrine growth factor amphiregulin in glioblastoma cells. Phosphatase and tensin homolog (PTEN) is a phosphatase which preferentially dephosphorylates phosphoinositide substrates and has been discussed to suppress glioblastoma cell proliferation by inhibiting the phosphoinositide 3-kinase (PI3K) pathway. Mutations and deletions of the PTEN gene were frequently observed in glioblastomas. Therapeutic overexpression of PTEN has been considered as a potential way to manage glioblastoma progression. Methods: PTEN was transiently overexpressed in the PTEN-deficient human glioblastoma cell line U87MG. The expression of HIF-2α and amphiregulin was studied using quantitative PCR. Results: We found that PTEN up-regulates the gene expression of HIF-2α and amphiregulin by suppressing the antioxidant effect of HIF-1α, another member of the HIF family of transcription factors. HIF-1α knockdown and antioxidant treatment represent potent ways to attenuate PTEN-induced HIF-2α up-regulation. Conclusions: Our results indicate that besides inhibiting PI3K pathway-dependent glioblastoma cell proliferation, PTEN also has the potential to promote HIF-2α-dependent glioblastoma growth. When using PTEN-overexpression for glioblastoma therapy, it would be necessary to take this biology into consideration. Disclosure: No conflict of interest disclosed. P818

Lichen substances sensitize the human glioblastoma cell line U87MG for the cytotoxic action of temozolomide Shcherbakova A.1,2, Koptina A.3, Ulrich-Merzenich G.2 Division of Biotechnology, Volga State Technical University, Yoshkar-Ola, Russian Federation, 2Medizinische Klinik III, UKB, Bonn, Germany, 3Division of Pharmacognosy, Uppsala University, Uppsala, Sweden 1

Introduction: Lichens are symbiotic organisms consisting of a mycobiont and an algae or cyanobacterium. They possess about 600 unique chemicals. Usnic acid, one of them, showed a broad spectrum of pharmacological activities of which the antioxidant and cytotoxic activity have gained major attention. We characterized and screened lichen extracts and their fractions for their (+)-usnic acid (US) content and their cytotoxic activity alone and in combination with temozolomide (TMZ) in U-87 MG glioma cells and in human skin fibroblasts (HSKF). Method: The lichens Cladonia arbuscula (L.) Hoffm., Evernia prunastri (L.) Ac. And Usnea barbata (L.) Wigg. were collected in Mari El Republic of Russia. Chloroform-, hexane (Hex), dichlormethan (DCM) and acetonitrile (ACN) extracts were prepared. US contents and the total phenol content were determined. Further chemical characterization was performed by HPLC. The total antioxidant activity of the extracts was measured. Toxicity was evaluated in HSKF (24 hrs) and in U-87 glioblastoma cells (8 to 24 hrs) by resazurin assay for the extracts, US and TMZ alone and in combination. Cytotoxic synergy was assessed by the combination index (CI). Phosphorylation of ERK1/2 and AKT were measured in U-87 cells by Western blotting. Results: The US content of the crude extracts (%) of C.arbuscula, E.prunastri and U.barbata were 8.9 ± 1.2, 5.7 ± 0.5, 74.5 ± 8.6.respectively. Their antioxidative capacity differed and was unrelated to the US content. In

Abstracts


CONTENTS AUTHOR INDEX

HSKFs the IC50 were 29.3 µg/ml, 78.6 µg/ml, 44.2 µg/ml and 15.5 µg/ ml for the extracts of E.prunastri (DCM, ACN) and C.arbuscola (Hex, DCM) respectively. In U-87 cells IC50 values of the extracts ranged from 10.8 µg to 98.1 µg/ml; the IC50 for TMZ and US alone were >400 µM and >100 µM respectively. The combination of TMZ and E.prunastri (ACN) (8:1) reached strong cytotoxic synergy (CI: 0.39–0.21) at high effect levels (≥0.9) with lowest toxicity in HSKF. Phosphorylations of ERK1/2 and Akt were dose and time dependently reduced. The combination TMZ and US was less effective (CI:0.78–0.48; effect level (≥0.9). Conclusion: Lichen substances, other than US, can sensitize the glioblastoma cell line U87 for cytotoxic actions of TMZ in vitro. Further studies on these secondary metabolites may foster besides drug development insights into drug resistance mechanism to TMZ in glioblastoma cells. Acknowledgement: A.Shcherbakova is supported by the DAAD. Disclosure: No conflict of interest disclosed. P819

ADAMTS13 regulates neutrophil recruitment in a mouse model of invasive pulmonary aspergillosis Hasibeder A.1, Prüfer S.2, Ebner K.3, Reuter S.1, Stein P.3, Scharrer I.1, Stassen M.2, Schild H.2, Jurk K.3, Bosmann M.1,3, Radsak M.P.1 University Medical Center of the Johannes Gutenberg-University, IIIrd Dept. of Medicine, Mainz, Germany, 2University Medical Center of the Johannes Gutenberg-University, Institute for Immunology, Mainz, Germany, 3University Medical Center of the Johannes Gutenberg-University, Center for Thrombosis and Hemostasis, Mainz, Germany 1

Introduction: Von Willebrand factor (VWF) is secreted as an acute phase protein during inflammation. The main mechanism regulating the size and prothrombotic activity of VWF is the specific proteolytic activity of ADAMTS-13. To determine the relevance of this regulatory pathway for the innate inflammatory response by polymorphonuclear neutrophils (PMN), we employed a mouse model of invasive pulmonary aspergillosis (IPA) where PMN functionality is crucial for fungal clearance and survival. Methods: IPA was induced by intratracheal application of Aspergillus fumigatus (A.f.) conidia in wildtype (129/Sv/Pas) or Adamts13 deficient (Adamts13–/–) mice. For the analysis of fungal load and histology, some mice were sacrificed 24 h after infection. Lung homogenates were either plated and cultured on Sabourough agar plates to assess fungal load as CFU. For histologic analyses, paraffin sections of the lungs were prepared and stained with mouse complement component C3d antibody. In addition, PMN functions, i.e. degranulation, oxidative burst activity, and CD62L shedding were assessed in PMN from wildtype and Adamts13–/– mice. Migration of purified human PMN isolated by dextran sedimentation and Histopaque® centrifugation towards A. fumigatus activated serum from wildtype and Adamts13–/– mice was evaluated (Corning® HTS Transwell®-96 well permeable support; 3µm). Results: While infected neutropenic mice developed lethal IPA, all wildtype mice survived the infection. Interestingly, Adamts13–/– mice displayed more severe signs of disease with a lethal course in about 24% of the animals. Examination of the lungs revealed a higher fungal burden along with increased signs of acute lung injury, complement deposition and levels of pro-inflammatory cytokines. Histology sections demonstrated a more pronounced perivascular leukocyte infiltration in support of a dysregulated inflammatory response in Adamts13–/– mice. Importantly, we observed no general defect in the activation of neutrophil effector functions in response to TLR agonist or in PMN-mediated fungal killing of conidia or hyphae in vitro. However, we found that ADAMTS-13 activity regulated complement activation subsequently inhibiting PMN migration. Conclusion: The proteolytic regulation of VWF by ADAMTS-13 is an important mechanism to control PMN recruitment in acute inflammatory processes, such as fungal pneumonias. Disclosure: No conflict of interest disclosed.

Abstracts

P820

Case-based lectures for medical students in an oncology group practice Weide R.1, Feiten S.2, Heymanns J.1, Thomalla J.1, van Roye C.1, Köppler H.1 Praxisklinik für Hämatologie und Onkologie, Koblenz, Germany, 2Institut für Versorgungsforschung in der Onkologie, Koblenz, Germany 1

Introduction: Optimal medical education should combine theoretical sessions with practical applications. A prerequisite are experienced lecturers, small groups and a personal relationship between students and lecturers. Personal and financial shortages are key challenges for the German university system to accomplish these goals. External university lecturers could unburden the task of the universities. Methods: Prospective evaluation of the academic education of medical students from the Johannes-Gutenberg-University Mainz in a community-based oncology group practice 2004–2014 using a standardized questionnaire. Results: 471 medical students took part in the haematology training course. All students had the option to pass the final examination which qualified them to acquire the university certificate “haematology training course”. The students allocated very good opinion scores concerning practical relevance, success of learning, learning atmosphere and engagement of the lecturers. 99% of the students stated that they would recommend the course to other students. Conclusions: Parts of clinical training during medical education can be accomplished by external university lecturers in their institutions. Quality and effectiveness of the training is judged by the students as being high. Systematic incorporation of external university lecturers, who are allowed to deliver their training of medical students in their institutions, could improve medical education and unburden universities. Disclosure: No conflict of interest disclosed.

Posterdiskussion

Verschiedene solide Tumore P821

Rapid response to lenvatinib in a patient with metastatic radio-iodine refractory differentiated thyroid carcinoma (DTC) progressing after multiple prior treatments with multi-target tyrosine kinase inhibitors (TKI) Mischler K.1, Kneifel S.1, Cathomas R.1 Kantonsspital Graubünden, Chur, Switzerland

1

Introduction: Patients (pts) with metastatic DTC primarily receive radioactive treatment with iodine-131. Until recently further options were very limited in case of progressive radioiodine refractory disease. The TKI sorafenib demonstrated improved progression free survival (PFS) but a low response rate. Recent data from a phase III trial show that the TKI lenvatinib achieved high reponse rates and a large improvement of PFS in treatment naive pts and after one prior line of TKI. Little is known about response to lenvatinib in heavily pretreated pts. Case report: A 45-year old man was diagnosed in 8/2008 with DTC (follicular structured oncocytic thyroid carcinoma) and underwent total thyroidectomy (pT2) followed by iodine-131 treatment (2’960 MBq). 7 months later multiple bilateral lung metastases (mets) were found that showed no uptake after another course of high-dose iodine-131 (7’400 MBq). In 8/2009 treatment with sorafenib was initiated. A partial remission (PR) was achieved. Progressive disease (PD) in the lungs and mediastinal lymph nodes (LN) was noted 7/2011, at which point the patient received treatment with weekly paclitaxel and a PI3K inhibitor in the setting of a phase I trial. Owing to further PD after 12 weeks therapy was switched to pazopanib 11/ 2011. Stable disease (SD) was seen until 9/ 2013 when treatment was changed to vandetanib due to PD. Within 6 weeks extensive and symptomatic PD with dyspnea and hemoptysis due to extensive pul-

Oncol Res Treat 2015;38(suppl 5):1–270

251


CONTENTS AUTHOR INDEX

monary lymphangiosis and bulky mediastinal LN mets was noted. Now 6 cycles of cisplatin and doxorubicin from 10/2013–1/2014 were applied with improvement of symptoms and radiologically stable disease. Starting in 2/2014 the patient received sorafenib with some decline of thyreoglobulin (TG), but he developed further progression 9/2014. At this point the novel TKI lenvatinib was started. Within 2 weeks symptomatic improvement and size reduction of palpable cervical LN mets were found. TG dropped from 33,000 to 1,200 ng/ml and a CT scan showed tumor regression. Due to intolerable gastrointestinal side effects the dose was reduced in two steps from the starting dose of 24 mg/d to 20 mg/d and further to 14mg/d. Conclusions: Our case demonstrates that the multitarget TKI lenvatinib can achieve a clinically meaningful reponse even in a patient with multiple prior treatments including three different TKIs and two different chemotherapy regimes. Lenvatinib needs careful monitoring and appropriate dose adjustments. Disclosure: No conflict of interest disclosed. P822

Needs, symptoms, and outcome of patients with advanced head and neck cancer treated on a palliative care inpatient ward Schmitz M.1, Wolfram M.1, Busch C.-J.2, Knecht R.2, Bokemeyer C.1, Oechsle K.1 Universitätsklinikum Hamburg-Eppendorf, II. Medizinische Klinik und Poliklinik, Hamburg, Germany, 2Universitätsklinikum Hamburg-Eppendorf, Klinik und Poliklinik für Hals-, Nasen- und Ohrenheilkunde, Hamburg, Germany 1

Introduction: Knowledge about needs and problems of patients (pts) suffering from advanced cancer is increasing substantially within the last years in general, but little is known about the specific needs and symptoms as well as treatment and outcome of pts with advanced head and neck cancer (aHNC) when admitted to a palliative care inpatient ward. Methods: A total of 97 pts (74% male, 26% female) with aHNC and a median age of 64 years (range, 35–90) were admitted to the palliative care inpatient ward at the University Medical Center Hamburg in 2011–2014. Most frequent primary tumor locations were oropharynx 19%, oral cavity 14%, hypopharynx 12%, multiple locations 12%. Distant metastases were present in 65%, 35% suffered from locally advanced disease. Results: At admission 75% of pts presented with an ECOG performance status of 3 or 4. The most frequent moderate or severe symptoms were weakness in 87% of pts, tiredness 64%, anorexia 60%, pain 52%, exertion 49%, dyspnea 45%, anxiety 38%, and depression 36%. Needs of care were support in daily activities in 81%, organization of further care in 68%, and distressed relatives in 62%. Advance directives and health care proxies were present in only 26% and 28%, respectively. Tracheotomy had to be performed in 40% and in 58% nutrition was applied via a PEG tube. During inpatient palliative care, 60% of pts received acute pain therapy, treatment of other symptoms 86%, physiotherapy 72%, social care 73%, psychological care 66%, music therapy 52%, pastoral care 48%. Additional palliative radiotherapy was applied to 7%, 16% received blood transfusions, and malignant effusions were drained in 6% of patients. Median duration of inpatient palliative care was 9 days (range, 1–35). A total of 53% of pts died on the palliative care ward and received specialized end-of-life care. Only 18% could be discharged to home care while 29% were admitted to hospice or nursery homes. Conclusion: In pts with aHNC weakness, tiredness, anorexia, and pain represent the most frequent symptoms. These pts seem to be referred to specialized palliative care rather late. At admission they present with several needs of care and psychosocial support. Discharge to home care seems to be possible less frequent than in other tumor entities. These preliminary results will provide a basis to improve assessment and treatment in pts with aHNC. Disclosure: No conflict of interest disclosed.

252

Oncol Res Treat 2015;38(suppl 5):1–270

P823

Results of the 3rd interim analysis of BRAWO – breast cancer treatment with everolimus and exemestane for ER+ women Tesch H.1, Grischke E.-M.2, Fasching P.3, Decker T.4, Uleer C.5, Schneeweiss A.6, Förster F.7, Wimberger P.8, Kluth-Pepper B.9, Schubert J.9, Bloch W.10, Jackisch C.11, Schütz F.6, Lüftner D.12 Oncology Bethanien, Frankfurt / Main, Germany, 2University Tuebingen, Department of Obstetrics and Gynaecology, Tübingen, Germany, 3University Erlangen, Department of Obstetrics and Gynaecology, Erlangen, Germany, 4 Medical Center, Ravensburg, Germany, 5Medical Center, Hildesheim, Germany, 6University Heidelberg, Department of Obstetrics and Gynaecology, Heidelberg, Germany, 7Policlinic GmbH, Chemnitz, Germany, 8University Dresden, Department of Obstetrics and Gynaecology, Dresden, Germany, 9 Novartis Pharma GmbH, Nürnberg, Germany, 10University of Cologne, Cologne, Germany, 11Sana Klinikum Offenbach, Offenbach, Germany, 12Charité – University Medicine Berlin, Department of Hematology, Oncology, and Tumor Immunology, Berlin, Germany 1

Introduction: BRAWO is a large German non-interventional study aiming to collect data of 3000 patients (pts) with advanced or /metastatic, hormone-receptor-positive and HER2-negative breast cancer treated with everolimus (EVE) and exemestane (EXE) under routine condition at about 400 sites. Main objectives are to extend the knowledge on a) the impact of physical activity on efficiency efficacy and quality of life, b) prophylaxis and management of stomatitis in clinical routine, and c) the sequence of therapy, when EVE is used in daily clinical practice. We report data of the 3rd preplanned interim analysis (IA) which was defined to take place 18 months after the inclusion of the 500st patient into the documentation. Methods: The 3rd IA (data cut-off 08 Jan 2015) covers data of the first 1300 documented pts (pts) at 314 sites. Here we report data on efficiacy and safety with focus on respiratory adverse events (AE). Results: At time of data cut-off 71% pts had discontinued the study, 29% were still ongoing. The two main reasons for discontinuation of therapy with EVE/EXE were progression n = 510 (37.1%) and AEs n = 268 (19.5%). Baseline characteristics: Median age 66 years, ECOG 0: 49.2%; median time since primary diagnosis: 6.9 years, visceral metastases: 54.4%; bone only metastases 25.3%. 86.5% of the patients received recommendations regarding stomatitis prophylaxis. Median PFS was 7.1 months (6.5–8.0; 95% CI). Tab. 1.

Most common adverse events (>10%)

All grades

Grades 3/4

Stomatitis

39.0%

2.8%

Fatigue

14.6%

1.6%

Nausea

12.2%

1.6%

Diarrhea

12.1%

1.0%

Dyspnoea

11.3%

3.1%

Most common respiratory, thoracic and mediastinal AEs (>2%)

All grades

Grades 3/4

Dyspnoea

11.3%

3.1%

Cough

9.0%

0.6%

Epistaxis

5.1%

<0.1%

Pleural effusion

4.8%

2.0%

Pneumonitis

4.2%

1.4%

Tab. 2.

Additional baseline characteristics and data on respiratory adverse events will be presented. Conclusion: The results observed in clinical routine are consistent with previous data reported in controlled clinical trials. Interestingly, stomatitis and pneumonitis were observed in fewer patients than in BOLERO-2. This may be an indicator that implementation of prophylaxis measures

Abstracts


CONTENTS AUTHOR INDEX

and experience in therapy management may have an impact on adverse event incidence. Disclosure: Hans Tesch: Employment or Leadership Position: Established oncologist in the community oncology practice at the BethanienHospital; Advisory Role: Novartis, Roche a.o.; Financing of Scientific Research: Novartis, Roche o.a. Diana Lüftner: Financing of Scientific Research: Member of speakers´ bureau Novartis. P824

Efficacy and safety of eribulin in combination with capecitabine in patients with metastatic breast cancer: An open-label, phase II dose-confirmation study Savulsky C.1, Twelves C.2, Anthoney A.2, Yin S.3, Evans T.R.J.4 Eisai Ltd, Hatfield, United Kingdom, 2Leeds Institute of Cancer and Pathology and St. James’s Institute of Oncology, Leeds, United Kingdom, 3Eisai Inc, Woodcliff Lake, United States, 4Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom 1

Introduction: Two phase III trials have demonstrated the efficacy of eribulin monotherapy in patients with metastatic breast cancer (MBC) who have progressed after previous anthracycline and taxane chemotherapy. Capecitabine is also an effective agent in MBC patients. A phase Ib dose-escalation study with two dosing schedules of eribulin in combination with capecitabine determined the recommended phase II dose. This phase II, dose-confirmation study aimed to determine efficacy and safety of this combination in MBC patients. Interim data to April 2014 are provided; final results will be presented at the meeting. Methods: Female MBC patients with ≤3 previous chemotherapy regimens (including an anthracycline and a taxane, but excluding capecitabine), ECOG performance status (PS) ≤1, and adequate hematological, renal and hepatic function were eligible. Patients received 1.4 mg/m2 eribulin mesylate (equivalent to 1.23 mg/m2 eribulin [expressed as free base]) IV on days 1 and 8 plus oral capecitabine 1000 mg/m2 BID on days 1–14 (21-day cycles) until disease progression, unacceptable toxicity or death. Six-weekly disease assessments (CT scans) were performed. The primary outcome was objective response rate (ORR); secondary outcomes included clinical benefit rate (CBR), progression-free survival (PFS), and safety/tolerability. Results: 42 females (median age: 52.5 years [range: 32–74]; ECOG-PS of 0: n = 18 [42.9%]) were enrolled; patients received study drug as a 1st-line (23.8% [n = 10]), 2nd-line (45.2% [n = 19]) or ≥3rd-line (31.0% [n = 13]) chemotherapy. A median of 8 (range: 1–30) treatment cycles were administered, with 94.4% and 94.5% of eribulin and capecitabine doses given, respectively, as planned. ORR was 42.9% (2 [4/8%] CR and 16 [38.1%] PR), CBR was 57.1%, and median PFS was 7.1 months (95% CI: 4.4, 9.8). The most common grade 3/4 adverse events (AEs) were neutropenia (66.7%), leukopenia (14.3%), febrile neutropenia (7.1%) and anemia, fatigue, nausea and peripheral sensory neuropathy (each 4.8%). AEs of special interest included: hand-foot syndrome (26.2%), peripheral neuropathy (23.8%), and diarrhea (21.4%). AEs led to study drug dose interruption/reduction/ withdrawal in 47.6%/42.9%/2.4% of patients, respectively. Conclusions: Eribulin in combination with capecitabine is efficacious in patients with MBC, with a safety and tolerability profile consistent with previous data. Phase III studies of this combination in MBC patients are warranted. Disclosure: Claudio Savulsky: Employment or Leadership Position: Eisai Ltd Employe, Director Clinical Development, Oncology. T.R. Jeffry Evans: Advisory Role: I have received payment from Eisai for consultancy on one of their other compounds (E7080) on one occasion in 2014; Expert Testimony: My institution received payment for commercial – sponsored clinical research trials with this, and other, Eisai compound.

Abstracts

P825

The metastatic profile of breast cancer stem cells and circulating tumor cells Apostolou P.1, Toloudi M.1, Kalliara I.1, Papasotiriou I.1 Research Genetic Cancer Centre Ltd, Florina, Greece

1

Introduction: Breast cancer is the most frequent type of cancer in women. Even though great progress has been made in treatment, resistance to chemotherapy and metastasis are still major problems. The study of cancer stem cells (CSCs) and circulating tumor cells (CTCs) could help to understand the above mechanisms, as both cells are implicated on the above. The present study aims to identify the gene expression of genes correlated with metastasis in specific organs in breast CSCs and CTCs. Methods: Human established breast cancer stems cells (provided by CelProgen), as well as breast CSCs and CTCs derived from six different patients suffered from breast cancer have been used. Each cell line was validated by using molecular and cellular-based methods (flow cytometry-qPCR). RNA was extracted from each cell line and whole genome gene expression microarrays have been performed. The data were normalized according to a normal (non-cancerous) sample. The array data were then evaluated with qPCR by using specific primers. Genes that correlated with general metastasis (TGF-bR2, ITGB-4R, ITGB-5R, ITGB-6R), metastasis to brain (STAT3, CX3CR1, DSC-2), liver (CXCR4, TNFRSF11A, FAS, HGFR), lung (IGF2R, MAPK3, MAPK1) and pleura (CCR6, MSLN) were studied. The relative quantification was performed according to Livak method, by using 18SrRNA as housekeeping gene. All the reactions were performed in triplicates. Results: Regarding the genes that correlated with general metastasis, there was observed an over-expression in all breast CSCs for integrins, while the TGFb-R2 was over-expressed in all CTCs. On the other hand, an under-expression was noticed for all genes correlated with brain metastasis in CSCs, but CX3CR1 and STAT3 were over-expressed in CTCs. The gene expression of HGFR was higher for all CSCs and was under-expressed in the majority of CTCs. The rest genes implicated in metastasis to liver were under-expressed almost in all samples. Concerning the MAPK kinases, on CSCs the MAPK3 was over-expressed, while MAPK1 was under-expressed. On CTCs, there was an over-expression. Finally, MSLN was not expressed in CTCs but only in CSCs. Conclusions: CTCs have different profile according to genes related to the possible organ of metastases compared with the CSC. This is what in clinical reality is described as metastases that will respond to second or third lines of therapies. However metastases to the brain may be refractory due to the features of CSCs. Disclosure: No conflict of interest disclosed. P826

P53 overexpression and high tumour proliferation (ki67) are associated with an adverse outcome in ductal pancreatic adenocarcinoma (PDAC) with adjuvant gemcitabine treatment Striefler J.K.1, Sinn M.1, Denkert C.2, Pelzer U.1, Bischoff S.3, Sinn B.V.2, Bläker H.2, Riess H.1, Lohneis P.2, CONKO Studiengruppe Charité Universitätsmedizin Berlin, CC14 Med. Klinik m. S. HämatologieOnkologie, Berlin, Germany, 2Charité Universitätsmedizin Berlin, Institut für Pathologie, Berlin, Germany, 3CONKO Studiengruppe, Berlin, Germany 1

Introduction: KRAS, CDKN2A/P16, TP53 and SMAD4/DPC4 are the most frequently altered genes in PDAC. They -as well as ki67- can be easily detected by immunohistochemistry (IHC). Previous investigations of these well-known markers in PDAC resulted in conflicting results. Methods: CONKO-001, a prospective randomized phase III study, investigated the role of adjuvant gemcitabine (gem) as compared to observation (obs) only. Tissue samples of 162 patients were collected and analysed by IHC for the expression of p16, smad4/dpc4, p53 and ki67.

Oncol Res Treat 2015;38(suppl 5):1–270

253


CONTENTS AUTHOR INDEX

Cases with nuclear p16 expression >5% of tumour cells were regarded as positive. For p53 the overall intensity (negative, weak, moderate and strong staining) and percentage of stained tumour cells (0%, 1–10%, 10–50%, 50–80%, >80%) was determined for each tumour. Strong and consistent p53 expression, displayed by an immunoreactive score of > = 6 points was regarded as p53 positive. IHC labelling of dpc4/smad4 was scored as intact when > = 5% of tumour cells exhibited cytoplasmatic staining. Ki67 pos tumour cells were counted in 300 tumour cells of each tumour. Expression in > = 12.83% of tumour cells was regarded as high proliferation. Expression was correlated with disease free and overall survival (DFS, OS) estimated with Kaplan-Meier-Method. Results: Low proliferation/low ki67 index was associated with better DFS (13.11 vs. 10.32 months, p = 0.041) and OS (30.23 vs. 20.37 months; p = 0.042) in the overall study population. This effect was restricted to gem treated patients (DFS 20.24 vs. 13.08 months; p = 0.041; OS 41.56 vs. 21.48 months; p = 0.051), while for patients in the obs group no association with proliferation was found. High p53 was associated with worse DFS (8.51 vs. 12.85 months; p = 0.035) and OS (18.23 vs. 28.78 months; p = 0.033). This effect could not be confirmed on survival in the overall study population or subgroup analysis. P16 and smad4/dpc4 expression had no influence on DFS or OS in the patients and subgroups investigated. Conclusion: P53 expression and tumour cell proliferation (ki67) demonstrate a significant impact on survival in patients with PDAC after curatively intended resection. For ki67, this effect was restricted to patients who received adjuvant gem and may contribute to clinical decision making towards an individualized therapy. Disclosure: No conflict of interest disclosed. P827

Metastatic pancreatic carcinoma: Observational registry study on quality of life and molecular biology of patients receiving Nab-paclitaxel/Gemcitabine first line therapy (a study in progress report) zur Hausen G.1, Pauligk C.1, Reichart A.1, Hozaeel W.1, Aldaoud A.2, Schöttker B.3, Hofheinz R.-D.4, Springfeld C.5, Sahin U.6, Al-Batran S.-E.1 Krankenhaus Nordwest gGmbH, Institut für Klinisch-Onkologische Forschung, Frankfurt am Main, Germany, 2Forschungsgemeinschaft Dr. Aldaoud/Dr. Schwarzer mbH, Leipzig, Germany, 3Gemeinschaftspraxis Dr. R. Schlag/ Dr. B. Schöttker, Würzburg, Germany, 4Universitätsmedizin Mannheim, III. Medizinische Klinik, Mannheim, Germany, 5Universitätsklinik Heidelberg, Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany, 6 Translationale Onkologie an der Universitätsmedizin Mainz gGmbH, Mainz, Germany 1

Introduction: Current studies resulted in a superiority of a combination of nab-paclitaxel and gemcitabine vs. gemcitabine mono therapy. However, there is no sufficient data available yet on the quality of life (QoL) of patients under this combination therapy and there is a high need to identify predictive and prognostic markers. In the framework of a multicenter registry study (‘QoliXane’), detailed QoL-data are now being collected and compared to existing data on gemcitabine mono therapy. Moreover, paraffin embedded tissues are collected for molecular analysis. Methods: QoL will be assessed by the EORTC-QLQ-C30 questionnaire and an additional questionnaire focusing on patient worries regarding risk of progression and potential side effects. Primary endpoint is the proportion of patients with maintained Global Health Status/QoL at 3 months. Secondary endpoints are QoL at different time points, efficacy (also in special subgroups of patients with high bilirubin levels) and safety as well as the identification of predictive and prognostic factors. 600 patients shall be enrolled at 80 study sites. Patients with metastatic pancreatic cancer are enrolled if they had no prior treatment in the metastatic stage and a combined treatment with nab-paclitaxel/gemcitabine is planned. QoL and observational data will be collected once a month over a 6-month period. Tumor containing paraffin embedded tissues are collected.

254

Oncol Res Treat 2015;38(suppl 5):1–270

Results: Recruitment started in the 4th quarter 2014. A total of 75 patients could be recruited by April 2015. A descriptive interim analysis of all patients with at least 3 cycles will be conducted regarding patient characteristics and the additional questionnaire by September 2015. Conclusions: QoliXane is a trial in progress with limited effort for the local investigators and the large potential to close an information gap within a highly important setting. Disclosure: Gerrit zur Hausen: Financing of Scientific Research: Lectures ­(Celgene); Other Financial Relationships: Reisekosten (Celgene). Salah-Eddin Al-Batran: Financing of Scientific Research: Advisory role & Lectures (Celgene); Expert Testimony: Research grant (Celgene). P828

Differential somatostatin and CXCR4 chemokine receptor expression in hepatocellular and cholangiocellular carcinoma Kaemmerer D.1, Schindler R.2, Mußbach F.3, Dahmen U.3, Dirsch O.4, Sänger J.5, Schulz S.2, Lupp A.2 Zentralklinik Bad Berka, Klinik für Allgemein- und Viszeralchirurgie, Bad Berka, Germany, 2Universitätsklinikum Jena, Institut für Pharmakologie und Toxikologie, Jena, Germany, 3Universitätsklinikum Jena, Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Jena, Germany, 4Universitätsklinikum Jena, Institut für Pathologie, Jena, Germany, 5Labor für Pathologie und Zytologie, Bad Berka, Germany 1

Introduction: Hepatocellular (HCC) and cholangiocellular carcinoma (CCC) display an exceptionally poor prognosis and especially for diffuse, multifocal or metastatic tumors no efficient standard therapy is available up to now. Recently, somatostatin analogs have been evaluated for the treatment of these malignancies, however, with conflicting results. Besides the presence of the different somatostatin receptor (SSTR) subtypes, also an expression of the chemokine receptor CXCR4 has been discussed in HCC and CCC. Since comprehensive data are still lacking especially for CCC, the aim of the present study was to comparatively assess the SSTR subtype and CXCR4 expression in both tumor entities. Methods: The expression of the SSTR subtypes 1, 2A, 3, 4 and 5 and of the CXCR4 was evaluated in a total of 71 HCC and 27 CCC by means of immunohistochemistry using novel monoclonal rabbit anti-human SSTR or CXCR4 antibodies, respectively. The immunohistochemical stainings were evaluated by means of the Immunoreactive Score and correlated to clinical data. Results: Regarding HCC, the number of positive cases and the intensity of expression for the SSTRs and for the CXCR4 were only low. The CXCR4 was the most prominent receptor in HCC and was present in about 40% of the cases, followed by the SSTR5, the SSTR2 and the SSTR3, which could be detected in about 15%, 8% or 5% of the tumors, respectively. No SSTR1 or SSTR4 expression could be observed in HCC. In comparison to HCC, the SSTR and CXCR4 expression was much higher in CCC. Here, the CXCR4 was present in 60% and the SSTR1 in about 70% of the tumors. The SSTR2 and the SSTR5 were detected in about 30% or 10% of the samples, respectively. None of the CCC displayed an SSTR3 or SSTR4 expression. HCC and CCC significantly differed with respect to the intensity of expression of the SSTR1 and of the CXCR4. In both tumor entities a significant correlation between the expression intensities of the different SSTRs was seen, but no correlation between the SSTRs and the CXCR4. With the proliferation marker Ki-67, in HCC a significant correlation was seen for the SSTR3 and for the SSTR5 expression and in CCC for the SSTR2 and for the SSTR5 expression. Conclusions: CCC, but not HCC, may be well suited for both SSTR- and CXCR4-based diagnostics and treatment. However, in case of targeting the SSTRs, due to the high expression rate of the SSTR1 in CCC, pan-somatostatin analogs should be preferred over octreotide (and derivatives). Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

P829

Blockade of cyclin-dependent kinase 5 (CDK5) identified as a novel therapeutic approach in controlling melanoma cell motility, invasiveness and metastatic spread Bisht S.1, Nolting J.1, Schütte U.1, Haarmann J.1, Brossart P.1, Flaherty P.2, Feldmann G.1 University Clinic Bonn, Department of Internal Medicine, Bonn, Germany, Duquesne University, Mylan School of Pharmacy, Medicinal Chemistry, Pittsburgh, United States 1 2

Introduction: Despite tangible progress in recent years, the overall prognosis of metastatic malignant melanoma remains poor as of to date. Therefore, better understanding of molecular mechanisms underlying melanoma progression and metastasis as well as identification of potential druggable targets to prevent these processes are urgently required. Methods: Expression levels of CDK5 and its activator protein p35 in human melanoma cell lines and primary tissue samples were evaluated using western blot and immunohistochemical analyses. Effects of shRNA-mediated inducible knockdown of CDK5 as well as pharmacological inhibition of CDK5 activity were determined using MTS, modified Boyden chamber and soft agar assays. A possible influence on tumorogenicity and systemic metastases of melanoma cells was analyzed using in vivo orthotopic xenografts and experimental metastasis models. Gene expression profiling using microarrays was carried out to identify possible downstream targets of CDK5 signaling in melanoma. Results: We confirm aberrant CDK5 activation in primary and metastatic melanoma lesions by overexpression of its activator protein p35 in human melanoma tissue microarrays (n = 163). Moreover, shRNA mediated inducible knockdown of CDK5 as well as pharmacological inhibition of CDK5 activity, were found to cause marked inhibition in cell motility, invasiveness, colony formation and anchorage independent growth of melanoma cells. In vivo, CDK5-knockdown inhibited growth of orthotopic xenografts as well as formation of lung and liver colonies in xenogenic or syngenic murine melanoma models mimicking systemic metastases. CDK5-knockdown was accompanied by dephosphorylation and overexpression of caldesmon, and concomitant caldesmon-knockdown rescued cell motility and pro-invasive phenotype of melanoma cells. Conclusion: This study for the first time provides strong experimental evidence for an involvement of CDK5 activation in metastatic spread of malignant melanoma, possibly via modulation of caldesmon phosphorylation. With pharmacological CDK5 inhibitors currently already being developed for other indications and tested with respect to their pharmacokinetics and toxicity profiles, this might open up a viable opportunity for therapeutic intervention aimed at blocking melanoma metastasis in the near future. Disclosure: No conflict of interest disclosed. P830

The histone deacetylase inhibitor valproic acid inhibits growth of tumors overexpressing SKI in a melanoma mice model Frech M.1, Teichler S.1, Stabla K.1, Neubauer A.1 Department of Hematology, Oncology and Immunology, Philipps University of Marburg and University Hospital of Giessen and Marburg, Marburg, Germany 1

Introduction: The Ski protein is known as a repressor of the TGFβ signaling pathway and an essential part of a co-repressor complex recruiting histone deacetylases (HDAC) (Nomura, 1999; Nomura, 2004). SKI is further an oncogene overexpressed in different solid tumors and leukemia (Bonnon & Atanasoski, 2012). In melanoma, Ski induces tumor growth by inhibiting TGFβ signaling and inducing β-catenin signaling (Reed et al., 2005). Our group showed that the high SKI expression in -7/del7q acute myeloid leukemias leads to an increased recruiting of HDAC, inhibiting the retinoic acid induced myeloid differentiation. The effect is partially revertable by treatment with the HDAC inhibitor (HDACi) valproic acid

Abstracts

(VPA) (Ritter et al., 2006). Hence, HDACi could also provide a treatment option in melanoma with a high SKI expression. Methods: We transfected the SKI-non-expressing cells NW1539 with a SKI-expressing or empty vector to generate NW1539-SKI or NW1539mock, respectively. We performed MTT assays to determine viability of these cells treated with VPA in vitro. Rag2–/–γc–/– mice were s.c. co-implanted with the melanoma cells NW1539-mock and NW1539-SKI. Mice were randomly allocated to the VPA (n = 10) or control (n = 9) group. In the VPA group daily treatment (600 mg/kg body weight) started upon a tumor size of ~200 mm3 for three weeks. Tumor sizes and volumes were determined every 2–3 days. Tumor tissue was further used for immunostaining and PCR. Results: In vitro experiments with the NW1539 melanoma cells showed an increased proliferation of the SKI-expressing cells. In contrast to the mock cells, SKI-expressing cells depicted a strong decrease in their viability upon VPA treatment. Thereupon, we tested the effects of VPA on the melanoma cells NW1530 ± SKI in a co-transplantation mouse model in vivo. SKI expression was analysed via Western analyses. Tumors originating from the NW1539-SKI grew faster than the tumors of the mock cells. The relative volumes of the SKI-expressing tumors further decreased significantly in comparison to the control tumors upon VPA treatment of the mice. Thus, confirming the results of the in vitro experiments. Conclusion: We show that a high SKI expression is correlated with a decrease of tumor cell growth in melanoma treated with the HDACi VPA. Hence, VPA offers a new option in the treatment of melanoma overexpressing the SKI oncogene. Ski thereby may be used as a marker and target protein for the treatment of human melanoma with VPA. Disclosure: No conflict of interest disclosed. P831

Atypical fibroxanthoma, pleomorphic sarcoma or malignant melanoma? – a case presentation Potenberg J.1, Dombrowski-Lütcke M.2, Minck C.2, Reyher-Klein S.2 Ev. Waldkrankenhaus, Onkologisches Zentrum, Berlin, Germany, 2Ev. Waldkrankenhaus, Berlin, Germany 1

Introduction: Making the diagnosis of a malignant melanoma could be challenging, especially if the neoplasia is not black and the manifestation is not the skin. Here, we present the case of a 48 years-old lady suffering from a soft tissue tumour. Methods: In October 2012 the patient developed a skin tumour of the lower abdomen. The neoplasia was diagnosed as plexiform neurofibroma. Some months later the tumour recurred. The diagnosis was then atypical fibroxanthoma/undifferentiated sarcoma with giant cells. In November 2013 a node in the right axilla was found. The histology was interpreted as pleomorphic sarcoma with neurogenic differentiation, possibly a peripheral malignant nerve sheath tumour. CT scan showed multiple metastases in the lung. 6 cycles of doxorubicin and ifosfamide were given with the result of stable disease. In June 2014 the tumour in the right axilla and the metastases in both lungs were excised. The histology was interpreted as pleomorphic tumour, possibly metastases of a malignant peripheral nerve sheath tumour or of a malignant desmoplastic melanoma. In November the tumour in the right axilla returned. The biopsy revealed malignant cells with immunohistochemically expression of S100 protein in all and Melan A in some cells. The tumour tested negative for HMB-45 and MNF116. The diagnosis was changed to malignant desmoplastic melanoma. Mutation analysis showed no driver mutation such as BRAF or c-Kit. The tumour board recommended the use of ipilimumab. Results: Evidently there were difficulties to distinguish between atypical fibroxanthoma/ pleomorphic sarcoma, neurogenic sarcoma, peripheral nerve sheath tumour and desmoplastic malignant melanoma. Atypical fibroxanthomas originate from the dermis, are S100 negative and exceptionally metastasize. S100 is a marker for neural crest-derived tumours and melanomas. The detection of some cells with expression of Melan A was interpreted as in favour of a malignant melanoma. The different tu-

Oncol Res Treat 2015;38(suppl 5):1–270

255


CONTENTS AUTHOR INDEX

mour manifestations were diagnosed by different pathologists and also referred to specialized centres. The diagnosis of a malignant melanoma was favoured by two specialists, two specialists favoured the diagnosis of a neurogenic sarcoma. Conclusion: It is worth the effort to diagnose or exclude the presence of a malignant melanoma because there are now a wide range of therapeutic options to choose from. Disclosure: No conflict of interest disclosed.

Wissenschaftliches Symposium CLL II V832

Genomic abnormalities and management of CLL Stilgenbauer S.1 Universität, Ulm, Germany

1

Chronic lymphocytic leukemia (CLL) is usually diagnosed in asymptomatic patients with early stage disease. The standard management approach is careful observation irrespective of risk factors unless patients meet the International Workshop on CLL (IWCLL) criteria for “active disease” that requires treatment. The initial standard therapy for most patients combines an anti-CD20 antibody (rituximab, ofatumumab or obinutuzumab) with chemotherapy (fludarabine/cyclophosphamide (FC), bendamustine, or chlorambucil) depending on multiple factors including the physical fitness of the patient. However, patients with very-high risk CLL because of deletion of 17p13 deletion (17p-) and/or mutation of TP53 (17p-/TP53mut) have poor responses to chemoimmunotherapy and require alternative treatment regimens containing B-cell receptor (BCR) signaling pathway inhibitors. The BCR signaling pathway inhibitors (ibrutinib targeting Bruton’s tyrosine kinase (BTK) and idelalisib targeting phosphatidyl-inositol 3-Kinase delta (PI3Kd) respectively) are currently approved for the treatment of relapsed/refractory CLL and patients with 17p-/TP53mut CLL. These agents offer great efficacy, even in chemotherapy refractory CLL, with increased tolerability, safety and survival. Ongoing studies aim to determine the best therapy combinations with the aim of achieving long-term disease control with the possibility of developing curative regimen for some patients. Disclosure: No conflict of interest disclosed. V833

Treatment of CLL: State of the Art 2015 Eichhorst B.1, DCLLSG Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany

1

Treatment decisions in CLL have become challenging, because of the wide spectrum of available treatment options. These range from watch&wait to different chemoimmunotherapies (CIT) or to new targeted therapies with kinase inhibitors (KI) or small molecules. Moreover, allogeneic stem cell transplantation (allo HSCT) is also still an option in very high risk fit CLL patients (pts). Other immunotherapies will be soon available. Asymptomatic pts in early stage CLL (Binet stage A and B without treatment indication) should still undergo the watch&wait strategy. Clinical trials are currently investigating if pts with high risk disease may benefit from early treatment with KIs. Pts in advanced symptomatic stage should undergo careful risk assessment including evaluation of genetic risk factors (del(17p)/TP53 mutation, IGHV status), physical fitnes and risk of infections. Fludarabine, cyclophosphamide and rituximab (FCR) is the most effective treatment in CLL so far with long-lasting remissions in subgroups of pts. Fit pts without del(17p)/ TP53mut and without prior history of severe infection should receive FCR accompanied by antiinfective prophylaxis. Fit elderly pts with a previous history for severe infections should be considered for bendamustine plus R (BR). BR is associated

256

Oncol Res Treat 2015;38(suppl 5):1–270

with a lower incidence of myelotoxicities and severe infections, but yields shorter response duration in comparison to FCR. For less fit pts without del(17p)/TP53mut CIT based on chlorambucil (CLB) have become the new standard therapy. Two phase III studies have shown the superiority of CLB plus CD20-antibody versus CLB alone, one of them even for overall survival. Pts with del(17p)/TP53 mutation respond less frequently to CIT and have a shorter duration of response. With the Btk inhibitor ibrutinib and the PI3Kinase inhibitor idelalisib two KIs are available for frontline therapy of these pts. For fit very high risk pts allo HSCT in first remission should be discussed. Short duration of response and especially progression during treatment with KIs is associated with a very poor prognosis. Early relapsed pts should be offered new substances inside or outside clinical studies. For pts with non-response to KIs switching KI or treatment with the bcl2-inhibitor Abt199, which will soon also be available outside clinical studies, are the currently preferred treatment options as well as allo HCT. Clinical studies will have to define the optimal use of the new substances in the near future. Disclosure: Barbara Eichhorst: Advisory Role: Roche, Janssen, Gilead, ­Mundipharma; Financing of Scientific Research: Roche, Janssen, Gilead, ­Mundipharma, Celgene; Expert Testimony: Roche, Mundipharma. V834

Significance of MRD assessments: Ready for clinical application ? Böttcher S.1 Universitätsklinikum Schleswig-Holstein, Campus Kiel, II. Medizinische Klinik, Kiel, Germany 1

With the advent of multiple highly effective treatment options, minimal residual disease (MRD) measurements, i.e. the quantitative and sensitive assessment of residual cell levels after and during therapy, gained interest in CLL. Conceptually, MRD might be applied (1) as an endpoint in randomized clinical trials in order to identify the more efficacious treatment arm or (2) as a means to tailor treatment intensity according to response in individual patients. These possible applications are based on recent research that improved the standardization of the technologies for MRD quantification and proved the clinical significance of MRD. Guidelines of the iwCLL defined MRD negativity as less than 1 CLL in 10000 benign leukocytes (10–4, Hallek, Blood, 2008), thus facilitating the comparability of results between trials. Multicolor MRD flow cytometry and ASO IGH RQ-PCR are the currently available methods for MRD quantification in CLL. Both technologies have been internationally standardized and were shown to be equally suitable for MRD determinations with a sensitivity of 10–4. The prognostic significance of an MRD threshold of 10–4 for progression free survival has been shown in univariate analyses of 10 individual publications comprising a total of 1157 patients. Moreover, recent data from 3 series additionally identified MRD as an independent prognostic variable even when tested together with established risk features in CLL (e.g. cytogenetics, IGHV, TP53 and SF3B1 mutational status). Additional work allowed the integration of MRD levels and pre-therapeutic risk features to identify high risk patients for early progression after immunochemotherapy. A recent combined analysis of MRD data from DCLLSG CLL8 and CLL10 trials clearly showed the value of MRD testing in both partial and complete responders. There are also data supporting the prognostic significance of MRD for overall survival in certain clinical situations. Available data support the use of MRD as a surrogate end-point in randomized clinical trials that compare different induction therapies. While there is only indirect evidence on the utility of MRD-guided treatment intensification or de-escalation in CLL, this concept might also be tested in clinical trials. Prior to the availability of additional corroborating data proving a clinical benefit of MRD-based treatment modifications the use

Abstracts


CONTENTS AUTHOR INDEX

of MRD to guide treatment decisions in individual patients out-side clinical trials is discouraged. Disclosure: Sebastian Böttcher: Financing of Scientific Research: Roche, AbbVie; Expert Testimony: Roceh, AbbVie, Celgene; Other Financial Relationships: Reisekostenerstattung: Roche. V835

T-cell prolymphocytic leukemia – an update Hopfinger G.1, Herling M.2 Medical University of Vienna, Department of Internal Medicine I,Bone Marrow Transplantation, Vienna, Austria, 2University of Cologne, Laboratory of Lymphocyte Signaling and Oncoproteome, Center for Integrated Oncology (CIO) Köln-Bonn and Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany 1

T-cell prolymphocytic leukemia (T-PLL) constitutes the most frequent mature T-cell leukemia with an incidence of ≈2/1,000,000. An aggressive clinical course and resistance to conventional chemotherapy with a poor response rate of only 30–45% to alkylating agents or CHOP is usually seen. With the introduction of the monoclonal anti CD52 antibody alemtuzumab, the overall response rate (OOR) has markedly improved. In relapsed patients, Alemtuzumab as a single agent resulted in an OOR of 76% including 60% of complete remission (CR).[1] When used as first line therapy, ORR of 91% including 81% CR was observed. With the aim to improve clinical outcome, a prospective trial was conducted by the German CLL Study group using an induction therapy with fludarabine, cyclophosphamide and mitoxantrone (FMC) followed by alemtuzumab consolidation therapy in 9 relapsed and 16 chemonaive patients. The ORR was 68% after FMC; after alemtuzumab administrated i.v., the ORR raised to 92% with median progression-free and overall survival of 11.9 and 17.1 months, respectively.[2] A subsequent trial using FCM along with alemtuzumab s.c. in 12 untreated and 4 with pretreated patients with T-PLL followed by an alemtuzumab maintenance therapy, resulted in an ORR of 68% with only a CR of 25% which is in line of the administration of FMC alone[3]. As alemtuzumab administered subcutaneously did not improve response rate when given as single agent [4] or in combination with chemotherapy [3], the i.v. route remains the recommended choice for T-PLL. Several strategies have been pursued in order to improve the dismal outcome of T-PLL by introducing high-dose therapy followed either by autologous stem cell transplantation (ASCT) or allogeneic transplant (AlloTx) in T-PLL. However, despite improved outcome after high-dose therapy followed by ASCT or alloTx, long term outcome is compromised by either high relapse after ASCT or TRM after alloTx.[5] In conclusion, improvement of both ORR and OS remains a challenge as induction therapy remains unsatisfactory and long term survival even after high dose is poor. References: 1 Dearden CE. Blood. 2001. 2 Hopfinger G. Cancer 2013. 3 Pflug N. ASH 2014 #1999. 4 Dearden CE. Blood 2011. 5 Herling M. Eur J Haematol. 2015. Disclosure: Georg Hopfinger: Advisory Role: Roche, Takeda; Financing of Scientific Research: Takeda, Roche, Gilead, Janssen. Marco Herling: No conflict of interest disclosed.

Freier Vortrag Immuntherapie III V846

Bispecific NKG2D-CD16 and -CD3 fusion proteins for enhancing immune responses of NK and T cells against leukemia cells Körner S.1,2, Jung G.3, Kanz L.2, Grosse-Hovest L.4, Salih H.1,2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Department for Internal Medicine II, Tuebingen, Germany, 2 Eberhard Karls University, Department of Hematology and Oncology, Tuebingen, Germany, 3Eberhard Karls University, Department for Immunology, Tuebingen, Germany, 4Synimmune GmbH, Tuebingen, Germany 1

Monoclonal antibodies (mAbs) have by now become an established tool in therapy of many malignancies. The interaction of a mAb’s Fc portion with Fcγ receptors (FcγR) on immune effector cells is important for its efficacy, but often insufficient to potently induce antitumor immunity e.g. due to FcγR polymorphisms. Moreover, Fc parts of mAbs may bind to FcγRs expressed on non-cytotoxic cells (e.g., platelets and B cells) and interact with FcγRs that do not trigger cytotoxicity (e.g. CD16b on granulocytes). These shortcomings can be overcome by novel antibody formats like bispecific antibodies allowing for improved activation of a specific pool of effector cells. Here we report on the development and preclinical characterization of two bispecific fusion proteins that target ligands of the immunoreceptor NKG2D (NKG2DL) which are widely expressed on malignant cells but generally absent on healthy tissue. Our fusion proteins consist of the extracellular domain of NKG2D as targeting moiety for tumor-expressed NKG2DL fused to Fab-fragments of either an agonistic CD3 or CD16 antibody. Specific binding of these NKG2D-CD3 and NKG2D-CD16 constructs was confirmed using NKG2DL-transfectants with regard to their target arm and either NK cells or T cells with regard to their different effector parts. Dose titration assays revealed an increased affinity of NKG2D-CD16 to the FcγR on NK cells as compared to our previously described Fc-optimized NKG2D-IgG1 fusion protein (e.g., Steinbacher et al, 2014), which was mirrored by a potently increased ability to induce lysis of NKG2DL-tranfectants and NKG2DL-positive primary acute myeloid leukemia (AML) cells by allogeneic NK cells. The novel NKG2D-CD3 construct in turn was found to potently activate allogeneic and autologous CD4+ and CD8+ T cells. Next we comparatively analyzed the efficacy of T cells and NK cells to lyse autologous leukemia cells upon treatment with NKG2D-CD3 and NKG2D-CD16, respectively, by using PBMC of AML patients (blast counts 30–70%) directly ex vivo. NKG2D-CD16 potently induced AML cell lysis, and this was, in line with their higher effector potential, by far exceeded upon stimulation of T cells with NKG2D-CD3. Taken together, we here introduce novel “antibody-like” bispecific constructs that take advantage of the highly tumor-restricted expression of NKG2DL and potently activate the reactivity of NK or T cells for immunotherapy of leukemia. Disclosure: No conflict of interest disclosed. V847

Side effects and cellular changes in the eye during therapy with epidermal growth factor receptor inhibitors Grewing V.1, Brandauer K.1, Mackensen F.1, Potthoff K.2 Universitäts-Augenklinik, Heidelberg, Germany, 2Medizinische Onkologie und Klinik für Strahlentherapie, NCT Heidelberg, Universitätsklinikum, Heidelberg, Germany 1

Introduction: Epidermal growth factor receptor inhibitors (EGFR-I) play a growing role in anticancer treatment. Side effects include skin reactions, mucositis as well as toxic effects in the eyes. Data concerning incidence, pathophysiology and exact manifestation of the ocular side effects are rare.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

257


CONTENTS AUTHOR INDEX

The aim of the study was to evaluate patients receiving EGFR-I treatment in terms of subjective eye discomfort and objective ocular findings including confocal microscopy of corneal epithelium and conjunctival goblet cells as the site of EGFR production. Methods: For analysis of subjective and objective eye discomfort the incidence of painful eye discomfort (VAS > 5), the time until first occurrence of eye discomfort, slit lamp examination (incl. Lid-parallel conjunctival folds (LIPCOFs), Break-up-time (BUT), Fluoresceine staining), Schirmer test and measurement of corneal epithelial and conjunctival goblet cell density with the Rostock-Cornea-Module (RCM, Heidelberg Eng.) were investigated. Results: 148 patients undergoing EGFR-I treatment (cetuximab n = 110, panitumumab n = 36, erlotinib n = 2) were questioned, 27/148 (18%) had a beginning of ocular discomfort (VAS > 5) after a mean of 7.3 ± 8.5 weeks since begin of EGFR-I treatment. 16/27 patients could be examined. All 16 patients complained of dry eye symptoms. Exam of the patients revealed clinical dry eye in 81% (13/16) of patients, 56% trichomegaly (9/16), 13% trichiasis (2/16), 56% blepharitis (9/16). The slit lamp examination showed reduced BUT (12/16), LIPCOFs (16/16), Meibomian gland dysfunction (16/16), keratitis (12/16), and corneal erosion (1/16). The density of corneal epithelium cells was negatively correlated to the duration of EGFR-I treatment (Spearman-correlation: rs = –0.6676, p = 0.0059). The conjunctival goblet cell density was with 54 cells/mm2 reduced in comparison to a normal cohort (unpaired t-test: p = 0.0061). Conclusions: Ocular discomfort is with an incidence of 18% a clinical relevant side effect during EGFR-I therapy. A wide spectrum of ocular side effects from dry eye-symptoms to keratitis up to corneal erosion could be observed. To our knowledge this was the first time that patients on EGFR-I treatment were examined with confocal microscopy. A reduction of corneal epithelial cell and conjunctival goblet cell density correlating with the EGFR targeted agent and the duration of treatment could be observed. Disclosure: No conflict of interest disclosed. V848

Combining T helper- and NKT-cell-mediated dendritic cell licensing enhances anti-tumor immune responses Heine A.1, Flores C.2, Held M.2, Brossart P.1, Kurts C.2 Medical Clinic III for Oncology, Hematology and Rheumatology, Bonn, Germany, 2Institute of Experimental Immunology, University of Bonn, Germany, Bonn, Germany 1

Purpose & Objective: Dendritic cell (DC) licensing is a requisite for the induction of efficient CD8+ T cell responses to extracellular antigens. Classically, DCs are licensed by CD4+ helper (Th) cells. As an alternative, NKT cells can replace CD4+ help. We have shown previously that combining both licensing systems results in synergistically enhanced CD8+ T cell responses, by mechanisms involving the chemokine receptors CCR4 and CCR5. Here we asked whether this chemokine synergism also improves anti-tumor immune responses. Materials and methods: We performed in vivo studies using two different tumor mouse models (B16-OVA and panc-OVA). Th-mediated DC licensing was initiated by the application of the TLR-9-ligand CpG, whereas NKT-cell-mediated DC licensing was induced by the NKT-cell-ligand α-Galactosylceramide (aGC). Results: After applying the model antigen Ovalbumin (OVA) together with both CpG and aGC, we detected higher amounts of antigen-specific CD8+ T cells that produced more pro-inflammatory cytokines and expressed more KLRG-1 than when only one licensing system was engaged. The combination of OVA/aGC/CpG resulted in higher antigen-specific T cell cytotoxicity in vivo, as well as enhanced effector T cell infiltration in the tumor. Tumors in these mice grew slower, showed reduced metastasis and overall survival was significantly improved. Conclusions: Combining classical and NKT-cell-mediated DC licensing allowed improved induction of tumor-specific CD8+ T cells leading to reduced tumor growth. These findings provide a promising approach of novel quality for designing better vaccines against tumors, since this com-

258

Oncol Res Treat 2015;38(suppl 5):1–270

bination not only enhances costimulatory molecules and cytokine production, but also engages different chemokine systems. Disclosure: No conflict of interest disclosed. V849

qPCR-based monitoring of hematopoietic chimerism after stem cell transplantation Ahci M.1, Crivello P.1, Stempelmann K.1, Buttkereit U.2, Shayegi N.2, Heinold A.3, Heinemann F.M.3, Horn P.A.3, Fleischhauer K.1, Beelen D.W.2 Institut für Zelltherapeutische Forschung/Universitätsklinikum, Essen, Germany, 2Klinik für Knochenmarktransplantation/Universitätsklinikum, Essen, Germany, 3Institut für Transfusionsmedizin/Universitätsklinikum, Essen, Germany 1

Introduction: Although short tandem repeat (STR) analysis has long been considered as the gold standard for monitoring of donor-recipient hematopoietic chimerism (HC) after allogeneic stem cell transplantation (SCT), interest in methods based on quantitative PCR (qPCR) for this purpose is increasing. This is not only due to the higher sensitivity of the latter approach, but also to its potential use for early detection of leukemia relapse with selective loss of mismatched HLA, a frequent condition after haploidentical SCT. Material and methods: We have performed a retrospective analysis of donor-recipient HC in a cohort of 30 patients who underwent HSCT from a 10/10 or 9/10 HLA-matched unrelated donor at the Department of Bone Marrow Transplantation at the University Hospital Duisburg-Essen, between the years 2006 and 2013, for the cure of AML, ALL, MDS or CMML. A total number of 482 follow-up samples, including 380 peripheral blood (PB) and 102 bone marrow (BM), were tested for HC in parallel by STR or by a commercial insertion-deletion qPCR (AlleleSEQR Abbott Molecular), with a threshold for positivity in qPCR HC of 0.1% for PB and 1% for BM. Results: For all 30 donor-recipient pairs, at least three informative markers (i.e. positive in the patient but negative in the donor) were available, with a median of 7 markers per patient. 2 markers selected for each patient showed high inter-marker reproducibility (r2 = 0.98). 88 samples drawn in parallel from BM or PB showed high levels of concordance between the two sources (r2 = 0.98). In 35 BM samples from 15 patients who maintained a stable complete chimerism (CC) in STR after transplantation, qPCR HC never exceeded the 1% threshold for positivity in BM, resulting in 100% specificity. In contrast, qPCR HC exceeding the 0.1% threshold for positivity in PB was found in 12/190 PB samples from patients with CC, resulting in a specificity of 93.7%. When at least 2 PB or 1 BM sample was available prior to relapse, at least one positive result was obtained for 7/7 patients by qPCR on PB, compared to 3/7 patients each for STR or qPCR on BM. Conclusion: HC monitoring after allogeneic SCT by qPCR on PB shows appealing sensitivity for relapse which is only partly traded off by decreased specificity. Sensitive qPCR targeting of inside HLA markers is under development to allow timely for detection of HLA loss relapse in mixed chimerism after SCT. Disclosure: Müberra Ahci: Expert Testimony: Bereitstellung der für das Forschungsvorhaben notwendigen kommerziellen kits durch die Firma Abbott Molecular. Dietrich Beelen: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V850

Induction of NK cell reactivity against myeloid leukemia by a novel Fc-optimized CD133 antibody Rothfelder K.1,2, Koerner S.1,2, Leibold J.3, Kousis P.1,2, Buehring H.-J.2, Haen S.2, Kuebler A.4, Andre M.4, Jung G.3, Kanz L.2, Grosse-Hovest L.5, Salih H.R.1,2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Department for Internal Medicine II, Eberhard Karls University, Tuebingen, Germany, 2Department of Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany, 3Department for Immunology, Eberhard Karls University, Tuebingen, Germany, 4Department of Pediatric Hematology and Oncology, University Children’s Hospital, Tuebingen, Germany, 5Synimmune GmbH, Tuebingen, Germany 1

NK cells largely contribute to the success of monoclonal antibody (mAb) application in cancer due to their ability to mediate antibody-dependent cellular cytotoxicity (ADCC), a feature considered critical for therapeutic efficacy. Up to now, no immunotherapeutic antibodies are available for the treatment of myeloid leukemias. Recently, we reported on the development of mAbs targeting CD133, which is expressed on a wide variety of tumor cells. Here we extend our analyses and provide data on the preclinical characterization of an Fc-engineered CD133 mAb for the treatment of myeloid leukemia. Compared to two other anti-human CD133 mAbs (clones AC133 and W6B3), which both bound to primary AML and CML cells in 15/25 and 5/6 cases, respectively, clone 293C3 recognized the leukemic cells in 22/25 AML cases and 5/6 CML cases. Based on these results, clone 293C3 was chosen to generate chimeric mAb with either a wildtype Fc part (293C3-WT) or a variant containing amino acid exchanges (S239D/I332E) to enhance affinity to the activating Fc receptor CD16 on NK cells (293C3-SDIE). Treatment with 293C3-SDIE resulted in significantly enhanced activation, degranulation and lysis of primary CD133-positive AML cells by NK cells in allogeneic and autologous experimental in vitro settings as compared to its wildtype counterpart. In NSG mice, induction of ADCC by treatment with 293C3-SDIE resulted in the elimination of patient AML cells by NK cells from a matched human donor. Considering the expression of CD133 on healthy hematopoietic progenitor cells, we further performed colony forming unit assays with healthy bone marrow cells. In line with the observed lower expression levels of CD133 on healthy compared to malignant hematopoietic cells, no relevant toxicity of 293C3-SDIE at the level of committed hematopoietic progenitor cells was observed. Thus, 293C3-SDIE constitutes an attractive immunotherapeutic compound for the elimination of minimal residual disease in CD133 bearing leukemia, especially in the context of allogenic SCT. Disclosure: No conflict of interest disclosed. V851

Hemophagocytic Lymphohistiocytosis (HLH) in adults: Results of the German HLH registry Heinevetter B.1, Schenk T.1, Brunkhorst F.M.2, Maschmeyer G.3, Rothmann F.3, Weber T.4, Müller M.5, Panse J.6, Janka G.7, Lehmberg K.7, Hochhaus A.1, La Rosée P.1

diagnostic criteria for adult patients (aHLH) often delays diagnosis. Since data on aHLH in Germany are not available, a national multicenter registry was initiated (www.hlh-registry.org). Methods: Patients (pts) with suspected aHLH were recruited from 19 institutions. To diagnose HLH, five out of eight pediatric HLH-2004 criteria had to be fulfilled: (1) fever, (2) splenomegaly, (3) cytopenia, (4) hypertriglyceridemia and/or hypofibrinogenemia, (5) hyperferritinemia, (6) high levels of soluble CD25, (7) low or absent NK-cell-activity and (8) hemophagocytosis. HLH diagnosis was validated by using the diagnostic H-Score (http://saintantoine.aphp.fr/score/). Clinical characteristics, laboratory findings, treatment and outcomes were collected. Results: From August 2010 to April 2015, 76 pts (33 female), median age 49 (17–81) years were enrolled. 57 pts fulfilled the HLH-2004 criteria. 52/57 pts achieved a probability of having HLH higher than 90% using the H-Score. Trigger diseases were neoplasia in 19/57 pts (lymphoma n = 14, acute leukemia n = 4, other malignoma n = 1), infections in 20/57 pts (EBV: 14/20). In 4/14 pts, EBV-HLH developed after allogeneic stem cell transplantation (allo-SCT) for AML (n = 2) and severe aplastic anemia (n = 2). Two pts were co-infected with EBV and H1N1 or HIV respectively. Other viral triggers were HIV, Parvovirus B19, HHV8 and CMV. 10/57 pts developed acquired HLH due to AID. The underlying trigger could not be identified in 8 cases. In 2 pts with infection-associated HLH and 1 pt with unknown trigger genotyping revealed hereditary HLH. Median time from symptom onset to diagnosis was 14 (1–90) days. 35/57 pts (61%) were alive after a median follow up of 56 (4–1984) days. 37/57 pts were treated with corticosteroids, 22/57 pts with etoposide and 22/57 pts received i.v. immunoglobulins. Allo-SCT was performed in 1/57 pts. Conclusions: Our pilot data suggest that trigger diseases are in accordance with the literature, with infections and neoplasia among the main causes. Diagnostic vigilance in all medical disciplines is required to diagnose aHLH prior to multiple organ failure development. We encourage nation-wide participation in the aHLH-registry in order to better understand the evolution and pathophysiology of this neglected disease. Disclosure: No conflict of interest disclosed.

Freier Vortrag

MDS experimentell V852

The human GFI136N variant is a novel prognostic marker that offers potential for personalized treatment approach for Myelodysplastic (MDS) and Acute Myeloid Leukemia (AML) patients Botezatu L.1, Makishima H.2, Michel L.1, Hoenes J.M.1, Vassen L.1, Vadnais C.3, Helness A.4, Marneth A.5, van der Reijden B.5, Radiovoyevich T.4, Germing U.6, Hass R.6, Schroeder T.6, Platzbecker U.7, Ehninger G.7, Lindberg E.8, Cazzola M.9, Boultwood J.10, Görgens A.11, Giebel B.11, Dührsen U.1, Maciejewski J.4, Moroy T.3, Khandanpour C.1

Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany, 2Zentrum für Klinische Studien, Universitätsklinikum, Jena, Germany, 3Klinikum Ernst von Bergmann, Klinik für Hämatologie, Onkologie und Palliativmedizin, Potsdam, Germany, 4Klinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum, Halle (Saale), Germany, 5Zentrum für Infektiologie und HIV, Vivantes AugusteViktoria-Klinikum, Berlin, Germany, 6Universitätsklinikum RWTH Aachen, Abteilung für Onkologie, Hämatologie und SZT, Aachen, Germany, 7Nationales Studienzentrum pädiatrische HLH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany

University Hospital Essen, Department of Hematology, Essen, Germany, Cleveland Clinic Taussig Cancer Institute, Translational Hematology and Oncology Research, Cleveland, United States, 3Institut de recherches cliniques de Montréal (IRCM), Hematopoiesis and cancer, Montreal, Canada, 4Cleveland Clinic, Cleveland, United States, 5Radboud University Nijmegen Medical Centre, Department of Laboratory Medicine, Nijmegen, Netherlands, 6University Hospital Duesseldorf, Department of Hematology and Oncology, Duesseldorf, Germany, 7University Hospital TU Dresden, Department of Internal Medicine I, Dresden, Germany, 8Karolinska University Hospital, Department of Medicine, Srockholm, Sweden, 9University of Pavia, Department of Molecular Medicine, Pavia, Italy, 10University of Oxford, Nuffield Department of Clinical Laboratory Sciences, Oxford, United Kingdom, 11University Hospital Essen, Institute for Transfusion Medicine, Essen, Germany

Introduction: Hemophagocytic Lymphohistiocytosis (HLH) is a severe hyperinflammatory syndrome, triggered by infections, neoplasia and autoimmune/-inflammatory diseases (AID). However, the absence of valid

Growth factor independence 1 (GFI1) is a pivotal transcription factor for definitive hematopoiesis. A variant allele of GFI1 (GFI136N), where a serine is replaced by an asparagine at position 36, has been described to

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

1

1 2

259


CONTENTS AUTHOR INDEX

predispose to AML development. We explored whether GFI136N predisposes to MDS and promotes progression to AML. Characterization of about 600 MDS patient samples from Germany, Netherlands, USA, Great Britain and Australia revealed that 11–13% were heterozygous for GFI136N compared to 5–7% in the healthy population. Moreover, a significantly reduced leukemia-free survival and a more advanced stage of MDS at diagnosis was observed for GFI136N MDS patients compared to GFI136S patients, independent of age, sex or mutations of previously described genes. To elucidate the mechanism behind this, we used knock-in mice expressing human GFI136N and GFI136S instead of murine Gfi1. Presence of GFI136N accelerated expansion of cells and colony forming capacity of AML1-Eto-9a or MLL-AF9 expressing murine cells. In vivo, the presence of GFI136N increased incidence and accelerated onset of AML in two AML models (Inv 16 and MLLAF9) as well as in Nup98-HoxD13 MDS mouse model. Genes involved in stem cell and leukemic stem cell signaling were upregulated in GFI136N mouse AML and MDS patient’s samples. ChipSeq and RNASeq revealed a higher degree of H3K4 methylation and H3K9 acetylation in GFI136N leukemic cells. Furthermore, gene expression arrays of mouse and human Gfi136N samples revealed a failure of lysine (K)-specific demethylase 1 (LSD1) and histone deacetylase (HDAC) to induce epigenetic changes. Thus, we predict that therapy with HDAC inhibitors (HDACi), as done for certain MDS patients, would likely fail in GFI136N patients. In contrast, treatment with histone acetyltransferase inhibitors (HATi) would be more beneficial. To test this, we treated GFI136S or GFI136N leukemic murine and human primary cells with HDACi and HATi. GFI136N cells did not respond to HDACi but dramatically responded to HATi. Preliminary in vivo data show a tendency for prolonged survival and reduced tumor burden of mice transplanted with Gfi136N leukemic cells after HATi treatment, in contrast to HDACi therapy. Collectively, these findings demonstrate that GFI136N is a novel prognostic marker for MDS that predisposes to AML via epigenetic changes. Our results support the use of HATi for GFI136N MDS patients, enabling a more personalized treatment approach.

expression that proved insensitive to the treatment with ABT-199/-737 despite being collected from a high-risk patient. Conclusion: MCL-1 conveys resistance to ABT-199/-737 in primary MDS samples.

Disclosure: No conflict of interest disclosed. V853

MCL-1 conveys resistance to ABT-199 in primary MDS samples Jilg S.1, Reidel V.1, Müller-Thomas C.1, Höckendorf U.1, Huberle C.1, Peschel C.1, Oostendorp R.A.1, Götze K.S.1, Jost P.J.1 Technische Universität München, III. Med.Klinik des Klinikums rechts der Isar, München, Germany

Fig. 1. Elevated protein levels of MCL-1 convey resistance to ABT-737 or ABT-199 in primary MDS samples.

1

Introduction: Clinical progression to higher-risk disease in MDS is accompanied by an increased resistance to undergoing apoptosis likely due to changes in the expression of BCL-2 family members. We have observed in a large cohort of MDS/sAML patients (n = 124) and healthy controls (n = 57) that ABT-199/-737 is specifically toxic for BM cells from highrisk MDS/sAML patients. Low-risk MDS and healthy controls remained largely unaffected. However few high-risk samples were resistant to treatment. As the expression levels of BCL-2 family members are known to affect the sensitivity to the BCL-2/-XL/-W inhibitor ABT-737, or the BCL-2-selective inhibitor ABT-199, we examined their expression in primary MDS samples. Methods: To understand the differential sensitivity to ABT-737 and ABT199, we assessed the protein levels of BCL-2, BCL-xL and MCL-1 in a collection of primary human MDS BM samples by intracellular FACS. Results: We observed that high MCL-1 expression mediated resistance to ABT-737 and ABT-199 induced cell death. Furthermore BCL-xL expression mediated resistance to ABT-199 without affecting the efficacy of ABT-737. High-risk MDS samples mostly expressed low MCL-1 and responded better to ABT-737/-199 whereas low risk samples mostly expressed high levels of MCL-1 rendering those samples resistant to the ABT compounds. The critical role of MCL-1 in treatment resistance can be appreciated in individual high-risk MDS samples with elevated MCL-1

260

Oncol Res Treat 2015;38(suppl 5):1–270

Protein expression levels of BCL-2, BCL-xL and MCL-1 were measured by intracellular FACS before treating MDS cells with ABT-737 or ABT199 for 72 hours. Shown are isotype control (grey) and expression level of the indicated proteins (red) for each patient sample. Cells were gated for CD34 expression and stained for viability using Annexin-V and 7-AAD. Disclosure: No conflict of interest disclosed. V854

Comparison of conventional Sanger with next generation sequencing for the identification of mutations in MDS patients in daily laboratory practice Martin R.1, Ganster C.1, Rietkötter E.1, Halbsgut N.1, Salinas-Riester G.2, Haase D.1 Universitätsmedizin Göttingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany, 2Georg August Universität, DNA Microarray and Deep-Sequencing Facility, Göttingen, Germany 1

Introduction: Myelodysplastic syndromes (MDS) are diseases of the bone marrow, which are characterized by a dysfunction of hematopoiesis commonly resulting in peripheral cytopenias, dysplasia and an increased risk of acute myeloid leukaemia (AML) development. Acquired somatic mutations account for MDS formation and constitute important markers for diagnosis especially for patients displaying normal karyotypes in

Abstracts


CONTENTS AUTHOR INDEX

cytogenetic analyses. Currently only a limited subset of genes is routinely analyzed by Sanger sequencing and the analyses often fail to detect mutations. The application of next generation sequencing (NGS) based analysis promises a cost-efficient and comprehensive screening method for clonal mutations in large scale. To test the applicability and reliability of this method we applied deep sequencing for samples from MDS patients and compared the results to previously obtained data from Sanger sequencing. Methods: We analyzed five patients (1× RCUD, 2× RAEB-1, 2× RAEB2) with normal karyotype. Genomic DNA was extracted from bone marrow samples (n = 4) and immunomagnetically enriched CD34+ peripheral blood cells (n = 1). PCR products containing selected target regions within 17 genes implicated in MDS formation were analyzed by Sanger sequencing and additionally pooled and sequenced using a MiSeq system (Illumina). Alignments were performed with bwa against the human reference genome hg19. Variants were identified filtered and annotated using Picard, GATK and Annovar. Results: Analysis of the NGS data showed that virtually all reads map to the amplified regions with sufficient coverage for variant detection. Sanger sequencing revealed variations in the genes SRSF2, ASXL1 and CBL. We were able to recover these aberrations also with the NGS approach. Besides this, the high read coverage allowed detection of additional, subclonal variants in the TET2 gene, which were likely overlooked in the Sanger sequencing analysis. Conclusion: Our results demonstrate that data from NGS analysis are consistent with those obtained from Sanger sequencing systems. Beyond that NGS allows a more efficient detection of subclonal variations, enables the screening of large gene sets, along with moderate costs, low amounts of sample material and a highly streamlined data analysis. We conclude that the application of NGS based techniques will enhance the accuracy of MDS diagnosis and prognosis. Disclosure: No conflict of interest disclosed. V855

Can flow cytometry predict cytogenetic abnormalities in patients with MDS – a prospective analysis in 345 MDS patients? Oelschlaegel U.1, Mohr B.1, Kramer M.1, Thiede C.1, Bornhäuser M.1, Ehninger G.1, Platzbecker U.1 University Hospital Dresden, Dresden, Germany

1

Introduction: Recently, we described a specific cell surface immunophenotypic profile in MDS patients harbouring a deletion 5q and proposed flow cytometry (FCM) as a rapid tool for diagnostics and disease monitoring (Oelschlaegel et al. 2015). Now, we aimed to test whether other cytogenetic abnormalities in MDS are mirrored in a specific immunophenotype as well. Therefore, bone marrow (BM) of MDS patients with (n = 136) or without (n = 209) cytogenetic abnormalities were investigated using FCM. Data were compared to healthy BM (n = 50), and non-clonal cytopenias (n = 109) as further controls. Methods: A standardized 8-color lyse-stain-wash procedure was performed. Samples were measured on a FACS Canto II. Sensitivity and specificity of the developed score were assessed using ROC analysis. Thereby, a logistic regression model allowed for a differential weighting of the single immunophenotypic features. Results: In a first step, we established specific immunophenotypic profiles separating the above mentioned cytogenetically abnormal subgroups from healthy BM and non-clonal cytopenias with a high specificity (94– 100%) and sensitivity (37–100%). Next, we were able to separate the respective cytogenetic subgroups from all other MDS (specificity: 87–96%; sensitivity: 43–56%) as well. Finally, we aimed at building immunophenotypic profiles separating the cytogenetic subgroups from all MDS plus non-MDS samples ending up with the following profiles, del(7q)/-7: abn granulocyte maturation pattern and aberrant CD56 (specificity = 97%, sensitivity = 56%); +8: abn myPC (>%, >CD45), normal SSC, and aberrant CD56 expression of granulopoiesis (specificity = 97%, sensitivity = 54%);

Abstracts

normal karyotype: high Ogata/FCSS-score, normal% myPC, and normal CD36 expression on granulopoiesis (specificity = 89%, sensitivity = 42%). Conclusions: This study gives further insight into MDS biology by pointing at the association of cytogenetic abnormalities and specific antigen expression profiles determined by FCM. Although not as sensitive as our previously described immunophenotypic profile for del(5q) MDS but highly specific, we propose to further validate the results in an additional MDS cohort and to prove their usefulness for diagnostics as well as therapy monitoring purposes. Disclosure: No conflict of interest disclosed. V856

Cooperations between molecular and chromosomal mutations in MDS Ganster C.1, Rietkötter E.1, Shirneshan K.1, Haase D.1 Universitätsmedizin Göttingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany 1

Introduction: In myelodysplastic syndromes (MDS), cytogenetic analysis detects somatic genetic aberrations in up to 75% and molecular sequencing in up to 90% of patients. Many frequently molecular mutated genes are located in chromosomal regions frequently affected by gross cytogenetic changes. Knowledge about the cooperation between molecular and cytogenetic mutations, the significance of homozygous compared to heterozygous somatic abnormalities and mechanisms leading to loss of function is still limited in MDS. Therefore, we aimed to investigate possible associations between molecular and chromosomal mutations in MDS patients. Methods: Cytogenetic testing for somatic mutations was performed using chromosome banding analysis. Interphase fluorescence in situ hybridization (FISH), multicolor FISH and molecular karyotyping (SNP array analysis) were used to characterize selected cytogenetic aberrations in more detail. Molecular testing for somatic mutations in the 17 most frequently mutated genes in MDS was done by Sanger sequencing. Results: We identified 33 MDS patients with cytogenetic as well as molecular aberrations. 22 patients showed one cytogenetic abnormality, one patient had two abnormalities and ten patients had at least three chromosomal changes. One gene was mutated in 17 patients; two genes were mutated in 10 patients; three genes were mutated in five patients and one patient showed four mutated genes. We detected a cytogenetic deletion, an amplification or a copy number neutral loss of heterozygosity (CNLOH) and simultaneously a molecular mutated gene in the cytogenetically affected region in 8/33 (24%) patients. The gene EZH2 in 7q35-q36 was mutated in 3/10 (30%) patients with chromosome 7 abnormalities. Remarkably, only 1/4 (25%) patients with cytogenetically detectable del(17p) also showed a molecular mutation in TP53 on 17p13.1. Further regions affected by two abnormalities were an iso(17)(q10) together with a molecular SRSF2 mutation (17q25.2), a CN-LOH on 4q together with a molecular TET2 mutation (4q24), a CN-LOH on 11q together with a molecular CBL mutation (11q23.3) and a micro-deletion on 2p that affected both DNMT3A alleles on 2p23. Conclusion: These mutation patterns suggest individual pathomechanisms leading to homozygous loss of function. On the other hand, as can be assumed by our TP53 data, also heterozygous mutations seem to play a relevant role in MDS. Disclosure: No conflict of interest disclosed.

Oncol Res Treat 2015;38(suppl 5):1–270

261


CONTENTS AUTHOR INDEX

V857

Azacitidine directly modulates function of mesenchymal stromal cells to alter bone marrow niche compostion and suppress malignant hematopoietic progenitors in MDS Huberle C.1, Wenk C.1, Witham D.1, Garz A.-K.1, Weickert M.-T.1, Pagel C.1, Müller-Thomas C.1, Oostendorp R.1, Peschel C.1, Götze K.1 Technische Universität München, III. Medizinische Klinik, München, Germany

1

Introduction: Hematopoietic stem cells (HSC) reside in a specific niche where interaction with mesenchymal stromal cells (MSC) governs quiescence, proliferation and fate decisions of HSC. HSC in low-risk myelodysplastic syndromes (MDS) have been shown to reprogram MSC. Evidence also suggests that an altered stromal microenvironment in MDS contributes to ineffective hematopoiesis. Azacitidine (AZA) is the standard treatment for higher-risk MDS. AZA induces cytotoxicity to cancer cells and inhibits DNA methyltransferase, allowing reversal of epigenetically silenced genes to restore impaired hematopoiesis. Methods: We asked whether treatment with AZA exerts a direct effect on the stromal microenvironment in MDS. We examined the effect of AZA on primary MSC in vitro as well as functional interaction between MSC and HSC after AZA treatment. Primary MSC were isolated from bone marrow, cultured under defined conditions and used up to 4 passages. Identity of MSC was confirmed by presence of characteristic cell surface markers by flow cytometry. Results: MSC from MDS patients showed reduced proliferation and altered morphology compared to healthy MSC. Treatment with AZA had no effect on cell surface marker expression of MSC. Growth kinetics revealed decreased proliferative capacity of healthy MSC at 100 µM AZA while MDS-MSC were more sensitive, with decreased growth at 10 µM. AZA significantly altered the ability of MDS-MSC to differentiate towards the adipogenic lineage while osteogenic differentiation was not affected. Gene expression and secretion of epigenetically regulated cytokines osteopontin (OPN) and SDF-1 were significantly decreased upon AZA treatment of MDS-MSC. Co-culture experiments showed that AZA pretreatment of MSC led to increased apoptosis of MDS CD34+ progenitors and inhibited short-term and long-term hematopoietic colony formation. This effect was recapitulated by addition of neutralizing OPN antibody to untreated MSC. Finally, Notch gene expression was upregulated in MDS CD34+ cells but not in healthy progenitors after contact with AZA pretreated MSC. In conclusion, we show that AZA directly modulates MSC function and alters niche composition leading to suppression of hematopoietic progenitors in MDS. Our data provide evidence that epigenetic regulation of MSC plays a role in MDS and suggest an additional mode of action by which AZA exerts its therapeutic activity.

but is restricted by mostly advanced age of MF patients. Here, we explore whether anti-thymocyte globulin (ATG) for GvHD prevention may induce liver toxicity in MF patients typically presenting with extramedullary hematopoiesis. Methods: Liver function parameters were evaluated during conditioning with or without ATG, in patients undergoing allo-HSCT for MF (between 2001 and 2014, n = 28) or for other indications (control; between 2013 or 2014, n = 130) in Basel. For an additional case-control analysis, case (MF) and matched control pairs were analyzed. Furthermore, two independent cohorts with n = 24 and respectively n = 37 patients with MF and allo-HSCT treatment were analyzed in collaboration with the Medical Centers in Freiburg and Tübingen. Results: Addition of ATG to the conditioning regimen raised bilirubin in both MF and control patients (Fig 1.), while leaving liver enzymes grossly unaltered. However, MF patients were more susceptible to ATG-induced hyperbilirubinemia with 16/17 (96%) of Basel MF vs. 43/77 (56%) control patients showing elevation of bilirubin (p < 0.001). We observed similar differences in a subgroup of patients receiving myeloablative conditioning (p < 0.001) and in the case control analysis (p = 0.039). ATG-induced hyperbilirubinemia in MF was mostly self-limiting, as in 9/16 (56%) of cases, bilirubin levels normalized within 7 days. In the Freiburg cohort, all but two patients received either ATG or alemtuzumab for T-cell depletion. In the ATG group bilirubin elevation was more frequent in (7/7 vs. 9/15) and peak levels were higher (87.69 vs. 33.4 umol/l, p = 0.002). In 35 MF patients transplanted in Tübingen and treated with ATG, 30 (86%) showed elevated bilirubin during conditioning in line with the results in the other cohorts.

Disclosure: Christina Huberle: No conflict of interest disclosed. Katharina Götze: Financing of Scientific Research: Celgene GmbH.

Freier Vortrag

Allogene Transplantation klinisch II V858

Hyperbilirubinemia in patients with myelofibrosis undergoing allogeneic hematopoietic stem cell transplantation and antithymocyte globulin treatment Ecsedi M.1, Schmohl J.2, Zeiser R.3, Drexler B.1, Halter J.1, Medinger M.1, Duyster J.3, Kanz L.2, Passweg J.1, Finke J.3, Bethge W.2, Lengerke C.1,4

Fig. 1. Peak bilirubin Basel patients.

Conclusion: Our findings indicate that MF patients have a higher risk for hyperbilirubinemia during allo-HSCT conditioning, which is further enhanced by ATG. While the ATG-induced hyperbilirubinemia is self-limiting it may alter clearance of concomitantly applied drugs. Disclosure: No conflict of interest disclosed.

University Hospital Basel, Clinic for Hematology, Basel, Switzerland, 2Medical Center, University of Tübingen, Department of Hematology and Oncology, Tübingen, Germany, 3Freiburg University Medical Center, Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg, Germany, 4 University Hospital Basel, Department for Biomedicine, Basel, Switzerland 1

Introduction: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative treatment for myelofibrosis (MF),

262

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

V859

Prognostic significance of the European Leukemia Net classification of genetic risk in patients with acute myeloid leukemia undergoing allogeneic stem cell transplantation Hemmati P.1, Terwey T.1, Na I.-K.1, Jehn C.1, Vuong L.1, le Coutre P.1, Dörken B.1, Arnold R.1 Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany 1

Introduction: Genetic abnormalities are among the strongest predictors for relapse and overall outcome of patients with acute myeloid leukemia (AML). Recently, the European Leukemia Net (ELN) proposed a revised classification based on the presence or absence of specific cytogenetic and molecular aberrations. So far, the prognostic significance of this system has not been firmly evaluated in patients with AML undergoing allogeneic stem cell transplantation (alloSCT). Patients and methods: We retrospectively 274 patients with AML aged < 60 years transplanted at our center between 2004 and 2014. Genetic risk was categorized according to the ELN and the Southwest Oncology Goup/ Eastern Cooperative Oncology Group (SWOG/ECOG) classification systems. At the time of alloSCT, 162 patients were in first complete remission (CR1), 46 patients were transplanted in CR>1 and 66 patients had active disease. Standard myeloablative conditioning (MAC) was given in 106 patients, whereas 168 patients received reduced intensity conditioning (RIC) prior to alloSCT. Grafts were from either related (N = 85) or unrelated (matched: N = 137, mismatched: N = 52) donors. Results: The median follow-up was 32 months for the surviving patients. At 5 years after alloSCT, disease-free survival (DFS) and the cumulative incidence of relapse (CI-R) of the entire cohort was 50% and 34%, respectively. Patients with an adverse risk karyotype by ELN definition had significantly lower DFS as compared to patients with a favorable or an intermediate risk profile, i.e. 30% versus 56% for favorable, 52% for intermediate-1, or 59% for intermediate-2 patients (p = 0.0064). Correspondingly, the CI-R was highest in patients with an adverse genetic risk profile, i.e. 53% at 5 years. In contrast, patients with favorable, intermediate-1, or intermediate-2 risk disease had a CI-R of 15%, 40%, or 21% (p < 0.001). In the group of patients with an intermediate-1 risk profile, an adverse outcome (relapse) was related to the presence of a FLT3-ITD mutation, whereas the CI-R in intermediate-1 patients lacking a FLT3-ITD was similar to the those in the intermediate-2 group. Conclusions: Our results indicate that the ELN classification of genetic risk is highly predictive for relapse and survival of patients with AML undergoing alloSCT. The adverse outcome of patients in the intermediate-2 subgroup is related to presence of a FLT3-ITD mutation. Disclosure: No conflict of interest disclosed.

constitution of natural killer (NK) cells is of notable interest due to their known capability to induce GVL without GVHD. This analysis is a single center prospective study performed at the University Hospital Regensburg investigating the possible correlation between the regeneration of NK cell subsets and the incidence of acute GVHD (aGVHD) and vice versa during the first 200 days following alloSCT. Data were collected from 2009 to 2012, and 342 samples of 107 patients were analyzed by FACS with focus on immature CD56high, mature cytotoxic CD56dim NK cells, and the ratio between these two populations (CD56dim:CD56high). Statistical analysis was performed using both logistic and B-spline linear regression. While 45 (42.05%) patients did not develop any GVHD during the first 200 days, 62 (57.9%) patients developed aGVHD (grade 1 n = 22, grade 2 n = 21, grade 3 n = 14, grade 4 n = 5) in median at day 28 (11–182) after alloSCT. Interestingly, a lower number of CD56high cells correlated with aGVHD (p = 0.045). In the longitudinal analysis aGVHD showed an impact on immune reconstitution after alloSCT. There was a clear association between incidence of aGVHD and delayed expansion of the total NK cell population (p = 0.032), in particular the CD56high NK cells (p = 0.001). Remarkably, we observed a significant correlation between the severity of aGVHD and the lower recovery of CD56high NK cells during aGVHD. It is well known that NK cell reconstitution following alloSCT goes along with higher numbers of CD56high that further differentiate into cytotoxic CD56dimCD16high NK cells. In our study, aGVHD not only correlates with reduced numbers of total NK cells, but specifically with an impaired early expansion of CD56high NK cells. In sum, these data let suggest a negative impact of aGVHD on early NK cell immune reconstitution, maturation and NK subset distribution. In addition, monitoring of early NK cell reconstitution after SCT may help to identify patients at risk for the development of severe aGVHD. Disclosure: No conflict of interest disclosed. V861

Allogeneic hematopoietic cell transplantation as salvage treatment in patients with relapsed/refractory aggressive B-cell lymphoma – results of a single-center retrospective analysis Dörfel D.1,2, Haen S.1, Vogel W.1, Kanz L.1, Wirths S.1, Möhle R.1, Faul C.1, Bethge W.A.1 Universitätsklinik Tübingen, Innere Medizin II – Onkologie, Hämatologie, Klinische Immunologie, Rheumatologie und Pulmologie, Tübingen, Germany, 2 Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Tübingen, Germany 1

Allogeneic stem cell transplantation (alloSCT) often remains the only curative treatment for hematological disorders. However, its success is frequently limited by acute and chronic graft-versus-host disease (GVHD) causing significant morbidity and mortality. One of the major challenges of alloSCT is to reduce the incidence and severity of GVHD while boosting the graft-versus-leukemia effect (GVL). In the setting of alloSCT, re-

Introduction: Patients with relapsed or refractory high grade lymphoma have a dismal prognosis. Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative salvage treatment but data on outcome are limited. Methods: We retrospectively analyzed all adult patients with high-grade lymphoma undergoing allo-HCT at our center between 2002 and 2013. Results: We identified a total of 38 patients receiving allo-HCT during this period. Histologies were DLBCL (n = 18), grey zone lymphoma (n = 2), Richter’s transformation (n = 18) from either follicular lymphoma (n = 9), CLL (n = 8) or marginal zone lymphoma (n = 1). The median age at time of allo-HCT was 46 (range 28 to 68) years. 68% of patients had relapsed after autologous transplantation prior to allo-HCT. In 32%, allo-HCT was used as primary salvage strategy in refractory patients or after inadequate autologous stem cell mobilization. Prior to allo-HCT patients were treated with a median of 5 chemotherapeutic regimens (range 1 to 8) and 32% of patients had refractory disease, 26% PR, 37% CR, and 5% unknown status. Myeloablative conditioning regimens (MAC) were used in 46% of patients and 54% were treated with reduced intensity conditioning (RIC). The median age for patients receiving MAC and RIC was 44 years and 54 years, respectively. MAC consisted either of 12 Gy total body irradiation (TBI) and high-dose cyclophosphamide (n = 15), busulfan in combination with

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

V860

Bidirectional relation between acute GVHD and NK cell subset reconstitution following allogeneic stem cell transplantation Ullrich E.1,2,3, Bakthiar S.1, Salzmann-Manrique E.1, Bremm M.1, Gerstner S.3, Mackensen A.3, Bader P.1,4, Hoffmann P.5,6, Holler E.5,6, Edinger M.5,6, Wolff D.5,6 Childrens Hospital, Division for Stem Cell Transplantation and Immunologie, Frankfurt, Germany, 2LOEWE Center for Cell and Gene Therapy, Frankfurt, Germany, 3University Hospital Erlangen, Department of Internal Medicine 5, Erlangen, Germany, 4LOEWE Center for Cell an Gene Therapy, Frankfurt, Germany, 5University Hospital Regensburg, Department of Internal Medicine III, Regensburg, Germany, 6University of Regensburg, Regensburg Center for Interventional Immunology, Regensburg, Germany 1

263


CONTENTS AUTHOR INDEX

cyclophosphamide or thiotepa (n = 2), or etoposide, ifosfamide and carboplatin (n = 1). The RIC regimen consisted of fludarabine in combination with either melphalan (n = 13), thiotepa/melphalan (n = 2), treosulfan (n =2), cyclophosphamide/TBI (n =1) or TBI (n = 3). Peripheral blood stem cells were used as graft source. Donors were matched related ( n = 16), matched unrelated (n = 16), mismatched unrelated (n = 4) or haploidentical (n = 3). Median follow-up was 12 months (range 1 to 149 months) with a Kaplan Meier estimated overall survival (OS) for all patients of 53%, 47% and 36% at 1, 2, and 5 years. The non-relapse mortality (NRM) at day 100, 1 year and 3 years was 21%, 24% and 29%. Patients after RIC had a significantly better OS at 3 years than patients after MAC (51% vs 24%, p = 0.01). The cumulative incidence of acute GvHD ≥II°was 18% and of chronic GvHD 37% (limited 21%, extensive 16%). Conclusion: Allogeneic HCT is a potential curative treatment option even in advanced and refractory patients with a long term survival of 36%. Disclosure: No conflict of interest disclosed. V862

A single center retrospective analysis on the impact of colonization and infection with multidrug-resistant Pseudomonas aeruginosa on the outcome after allogeneic hematopoietic cell transplantation Heidenreich D.1, Kreil S.1, Nolte F.1, Reinwald M.1, Hofmann W.-K.1, Klein S.A.1 III. Medizinische Klinik, Universitätsmedizin, Mannheim, Germany

1

Introduction: Multidrug-resistant bacterial pathogens (MRP) such as vancomycin-resistant enterococci (VRE) and extended-spectrum beta-lactamase producing bacteria (ESBL) are an emerging challenge in allogeneic hematopoietic cell transplantation (HCT). Among gram-negative bacteria Pseudomonas aeruginosa has a pronounced pathogenicity. Thus, it has to be assumed that multidrug-resistant P. aeruginosa (MRPA) may cause relevant morbidity and mortality in HCT. It was the aim of this study to analyze the issue of MRPA in HCT. Patients and methods: A total number of 98 consecutive patients who received the first HCT from 2010 to 2014 were retrospectively analyzed. The underlying diseases were AML (55), ALL (7), MPN (10), MDS (14), lymphoma (8), and multiple myeloma (4). Conditioning was myeloablative in 29 and reduced intensity in 69 patients. Stem cell sources were peripheral blood (86) or bone marrow (12) from matched siblings (23), matched unrelated (58), mismatched (7) or haploidentical donors (10). As baseline investigation patients underwent a screening for MRP. Swabs from nose, throat, axilla, urethra and anus as well as stool and urine were collected. In addition routine microbiological investigations were performed whenever needed. ESBL were categorized as multi-resistant gram-negative bacteria (4MRGN, resistant to cephalosporins, piperacillin, fluorochinolones and carbapenems) or as 3MRGN (resistant to 3 of these 4 drug groups). Results: 8/98 patients were colonized by MRPA either at baseline [n = 2 (both 4MRGN)] or at any other time point until day 100 [n = 6 (2×3MRGN, 4×4MRGN)]. Out of these 8 patients 5 developed an infection with MRPA: 4x septicemia and 1x pneumonia with 4MRGN P. aeruginosa. All patients with 4MRGN P. aeruginosa septicemia died. Moreover, both patients who had been already colonized by 4MRGN P. aeruginosa at baseline died transplant related. Thus, NRM of MRPA colonized patients was 4 out of 8. In contrast to these dismal results, 2-year NRM of patients without MRP colonization and of patients who were colonized by non-MRPA MRP was low and essentially identical (18% and 17%). Discussion: Colonization with MRP other than MRPA had no negative impact on the NRM after allogeneic HCT. However, patients colonized by 4MRGN P. aeruginosa seem to have a dismal outcome. HCT of these patients should be considered with care. We therefore suggest including screening for MRP in the pretransplant work-up in order to identify patients colonized by MRPA. Disclosure: No conflict of interest disclosed.

264

Oncol Res Treat 2015;38(suppl 5):1–270

V863

Bone marrow harvesting in an outpatient setting is save and feasible in the majority of allogeneic bone marrow donors Lisenko K.1, Stadtherr P.1, Bruckner T.2, Pavel P.3, Heilig C.1, Schmitt A.1, Brandt J.1, Dreger P.1, Ho A.D.1, Witzens-Harig M.1, Wuchter P.1 Universitätsklinikum Heidelberg, Abteilung Innere Medizin V, Heidelberg, Germany, 2Universität Heidelberg, Institut für Medizinische Biometrie und Informatik, Heidelberg, Germany, 3Institut für Klinische Transfusion und Zelltherapie (IKTZ), Heidelberg, Germany 1

Introduction: Donors undergoing bone marrow (BM) harvest under general anesthesia have traditionally been admitted to the hospital for several days. However, there is a growing demand to perform this procedure in an outpatient setting for different reasons: Mainly the wish of the donor for a rather short stay in the hospital, but there is also an increased pressure on the accessibility of hospital beds in many centers. The aim of this retrospective study was to demonstrate the feasibility and safety of outpatient BM harvesting in allogeneic donors. Methods: Data of related and unrelated donors that underwent allogeneic BM harvest under general anesthesia at our institution on an out(n = 177) or an inpatient (n = 49) basis from 2002 to 2014 were analyzed retrospectively. To identify factors that predispose for hospital admission we compared donor-specific characteristics (sex, age, body weight [bw], foreign nationality [i.e. need for a professional interpreter], related/unrelated donor) and collection parameters (total nuclear cell number [TNC], operation time, total harvested BM volume, volume per kg body weight) between the out- and inpatient setting. Results: There were no significant differences in sex, age and donor’s body weight between the two subgroups. However, among inpatients the proportion of related donors and donors of foreign nationality requiring an interpreter was considerably higher compared to outpatients (p = 0.09 and p = 0.05, respectively). Furthermore, we found a significantly higher collection volume per kg bw of the donor in in- compared to outpatients (15.0 versus 12.8 ml/kg bw, p < 0.01). Conclusions: Our study demonstrates that performing allogeneic BM harvest in an outpatient setting is safe and feasible for the majority of donors. Besides socio-cultural factors, a high volume of collected BM per kg donor`s body weight represents a major indicator for inpatient admission and should be considered when planning for the procedure. Disclosure: Katharina Lisenko: No conflict of interest disclosed. Patrick Wuchter: Advisory Role: Consultancy, honoraria and membership on Advisory Boards of Sanofi-Aventis.

Wissenschaftliches Symposium PET bei Lymphomen V868

FDG-PET in lymphoma: Work in progress Gallamini A.1 A. Lacassagne Cancer Center, Research, Innovation and Statistics, Nice, France

1

The present review will try to demonstrate how FDG-PET integrated with Computed Tomography (FDG-PET/CT) is now considered an essential tool for lymphoma management. Beside its role lymphoma staging and restaging, which was settled in the millennium turnaround, PET scan became a very important tool for treatment monitoring both in first and second-line therapy and to guide consolidation radiotherapy in residual mass after chemotherapy. In the first part of the talk it will be shown how FDG-PET/CT has revolutionized the procedures for lymphoma staging and restaging, limiting the role of historical morphological imaging or invasive procedures such as contrast-enhanced CT scan and bone marrow trephine biopsy. The new rules for PET scan interpretation at baseline and for response assessment (the Lugano criteria) will be also presented and their integration with other diagnostic tests commented, as recently proposed in the expert consensus workshop during the 12th ICML meeting. A

Abstracts


CONTENTS AUTHOR INDEX

special emphasis will be used for the peculiar role of FDG-PET in follicular lymphoma staging and restaging and its relationship with minimal residual disease (MRD) monitoring during treatment. The second part of the talk will be focused on the issue of PET scan intended as surrogate test for chemo-sensitivity, and it will be shown how PET transformed from a simple imaging technique to a prognostic tool indissoluble from the Hodgkin Lymphoma (HL) therapeutic strategy, to predict the final treatment outcome. As a consequence, several PET response-adapted strategies have been proposed in clinical trials, some of them already concluded, while other are still ongoing. In early stage HL, two large international phase II trials, aimed at exploring the role of treatment de-escalation have been concluded and published while preliminary results several large prospective trials on the role of both therapy escalation and de-escalation in advanced stage HL and aggressive lymphoma are available. The most recent and modern aspect of prognostication in lymphoma will be presented, combining PET imaging with biomarkers, and tumour- and host-related prognostic factors. Finally, the new frontiers of PET scan interpretation with quantitative metrics (Q-PET) using the Standardized Uptake Value (SUV) and Metabolic Tumour Volume (MTV) will be presented, and the advantages over traditional qualitative PET scan interpretation discussed. Disclosure: No conflict of interest disclosed. V869

PET in aggressive lymphomas Hüttmann A.1 Universitätsklinikum Essen, Klinik für Hämatologie, Essen, Germany

1

Virtually all aggressive lymphomas can be visualized by positron emission tomography (PET) using the tracer 18-fluor-deoxyglucose. As a staging tool, PET offers the ability to resolve unclear computertomographic (CT) findings which may lead to up- or downstaging in some cases. However, in most instances a more accurate staging will not result in a change of treatment. From a more experimental point of view, staging lymphomas with PET alone is an interesting option as it may reduce radiation exposure and spare patients the nephrotoxic effects of contrast medium. During the course of lymphoma treatment it may be desirable to identify patients responding or not to standard chemotherapy. If performed under strict procedural conditions, the prognostic value of interim PET after two cycles of chemotherapy has been confirmed in several independent studies. If, in case of a favourable interim PET, standard treatment is continued, long term outcome is favourable, too. It is unclear, however, which alternative treatment strategies could lead to better outcome in patients with an unfavourable interim PET. Just as on interim PET, a normal finding on endof-treatment PET is reassuring for patients, because long-term prognosis is good. By contrast, how patients with end-of-treatment PET findings suggestive of persistent disease should be handled, has not been defined. They may be offered watchful waiting, further diagnostics (e.g. biopsy) or even therapy (e.g. radiation therapy or intensified chemotherapy). Managing asymptomatic patients with persistently abnormal PET findings is a clinical issue requiring further research. Disclosure: No conflict of interest disclosed.

Fortbildung

Kompetenznetzwerk Leukämien – neue Erkenntnisse bei AML/ALL/CML/MDS V872

MDS: Therapeutic implication of mutations Hofmann W.-K.1 Universitätsmedizin Mannheim, Hämatologie und Onkologie, Mannheim, Germany 1

Over the last decades, this diagnosis and classification of myelodysplastic syndrome (MDS) was supplemented by a number of chromosomal aberrations and molecular changes which are specific for MDS and correlate with distinct diagnostic and prognostic subtypes of this heterogeneous disease. New molecular high-throughput techniques such as single nucleotide polymorphism (SNP) arrays, gene expression arrays, DNA next generation sequencing and DNA-methylation arrays provide a way to further understand the pathogenesis of MDS. These technologies can give information on genetic alterations and changes in gene regulation even on the level of single genes (e. g. ASXL1, p53). Strong efforts to identify specific molecular genetic patterns that are associated with different risk-groups in MDS are underway. The aim is to further identify distinct subtypes in MDS for more effective risk assessment. So far, more detailed molecular diagnostic parameters available for patients with MDS have resulted in more reliable diagnosis and prognostic scoring. However, whether this can help to select MDS patients for specific therapies including allogeneic stem cell transplantation and therefore to improve clinical management of these patients is the major issue for recent and future translational approaches in MDS. Disclosure: No conflict of interest disclosed.

Freier Vortrag Prostatakarzinom V878

Marine compound rhizochalinin shows high anticancer activity in castration resistant and AR-V7 positive prostate cancer cell lines Dyshlovoy S.1,2,3, Otte K.1, Hauschild J.1, Venz S.4,5, Christ T.6, Bauer C.K.7, Bähring R.7, Amann K.8, Alsdorf W.1, Madanchi R.1, Schumacher U.9, Walther R.5, Makarieva T.2, Guzii A.2, Tabakmakher K.2, Kasheverov I.10, Kudryavtsev D.10, Stonik V.2, Bokemeyer C.1, Honecker F.1,11, von Amsberg G.1 University Medical Center Hamburg-Eppendorf, Department of Oncology, Haematology and Bone Marrow Transplantation, Section Pneumology, Hubertus Wald -Tumorzentrum, Hamburg, Germany, 2G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Vladivostok, Russian Federation, 3Far Eastern Federal University, Vladivostok, Russian Federation, 4Department of Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany, 5Interfacultary Institute of Genetics and Functional Genomics, Department of Functional Genomics, University of Greifswald, Greifswald, Germany, 6University Medical Center Hamburg-Eppendorf, Department of Experimental Pharmacology and Toxicology, Hamburg, Germany, 7University Medical Center Hamburg-Eppendorf, Department of Cellular and Integrative Physiology, Hamburg, Germany, 8University Medical Center Erlangen, Nephropathology Department, Erlangen, Germany, 9University Medical Center Hamburg-Eppendorf, Department of Anatomy and Experimental Morphology, Hamburg, Germany, 10Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Federation, 11Tumor and Breast Center ZeTuP St. Gallen, St.Gallen, Switzerland 1

Introduction: Despite recent progress in the treatment of castration-resistant prostate cancer (CRPC), development of resistance to current agents remains a challenge.

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

265


CONTENTS AUTHOR INDEX

We examined the efficacy of rhizochalinin (Riz) – a marine sphingolipid-like compound – in a human prostate cancer model. Methods: Anticancer activity of rhizochalinin was investigated using the CRPC cell lines PC3 and DU145 as well as the AR-V7 positive cell lines 22Rv1 and VCaP. Effects on cell viability and cell cycle were investigated by MTT, trypan blue staining and flow cytometry. Mode of action of the compound was analysed by fluorescence microscopy, Western blot analysis and a global proteome screening approach (2D-PAGE followed by mass spectrometry analysis). Synergy of rhizochalinin in combination with cabazitaxel was evaluated using the Chou-Talalay method. Results: In vitro Riz significantly decreased viability and inhibited proliferation of PC3, DU145, 22Rv1 and VCaP cells in a dose-dependent manner at low micromolar concentrations with 22Rv1 and VCaP showing the highest sensitivity towards the drug. Riz induced caspase-3-dependent apoptosis, down-regulation of the anti-apoptotic protein survivin, up-regulation of the pro-apoptotic factors p53, PTEN and p21 and a G2/M cell cycle arrest. Up-regulation of LC3BII, a marker of type II cell death, suggests that autophagy-associated processes are involved in the cellular response to Riz. The global proteome analysis revealed regulation of several proteins associated with formation of metastases, tumor cell invasion, and malignant growth. Among others, alteration of keratin 81, IL-1beta, and Hsp71, as well as down-regulation of LASP1 and stathmin were observed. A synergistic effect of Riz in combination with cabazitaxel was detected. Ongoing research includes electrophysiological experiments to study effects of Riz on ion channel function and membrane integrity as well as xenograft experiments to investigate the in vivo efficacy and toxicity of Riz. Conclusion: Rhizochalinin is a promising novel substance for the treatment of CRPC, showing high in vitro efficacy in castration resistant and AR-V7 positive prostate cancer cell lines. Regulation of pro-apoptotic as well as autophagy-associated pathways are supposed modes of action of cytotoxicity of the substance. Acknowledgement: This research is supported by the Eppendorfer Krebsund Leukämiehilfe, the Hamburger Krebsgesellschaft and the Erich and Gertrud Roggenbuck-Stiftung. The first and second author contributed equally. Disclosure: No conflict of interest disclosed. V879

Radium-223 in an international early access program (EAP): Effects of concomitant medication on overall survival in metastatic castration-resistant prostate cancer (mCRCP) patients Wirth M.1, Saad F.2, Galceran J.C.3, Gillessen S.4, Heinrich D.5, Gratt J.6, Miller K.7, Nilsson S.8, O Sullivan J.9, Tucci M.10, Heidenreich A.11 University Hospital Carl Gustav Carus, Dresden, Germany, 2University of Montreal Hospital Centers, Montreal, Canada, 3Vall Hebron University, Vall D`Hebron Institute of Oncology, Barcelona, Spain, 4Kantonsspital, St. Gallen, Switzerland, 5Akershus University Hospital, Lorenkog, Norway, 6Bayer HealthCare Pharmaceuticals, Whippany, United States, 7Charité University Medicine, Berlin, Germany, 8Karolinska University Hospital, Stockholm, Sweden, 9 The Centre for Cancer Research and Cell Biology, Queen`s University Belfast, and the Northern Ireland Cancer Centre, Belfast, Ireland, 10San Luigi Hospital, Orbassano, Italy, 11University Hospital, RWTH, Aachen, Germany 1

Background: The pivotal ALSYMPCA study reported improved overall survival (OS) in bone symptomatic mCRPC patients (pts) treated with radium-223 (Ra-223) vs placebo (median 14.9 vs 11.6 months [mos], HR = 0.70). Data from 696 EAP pts recruited from 14 countries (Europe, Canada, Israel) are presented. Methods: In this prospective phase IIIb study, mCRPC pts with symptomatic or asymptomatic bone metastases (no visceral disease) received Ra223 50 kBq/kg (iv injection) every 4 weeks for 6 cycles. Primary endpoints were safety and OS. The effects of concomitant medications, baseline (BL) pain, alkaline phosphatase (ALP) and ECOG PS on OS were assessed.

266

Oncol Res Treat 2015;38(suppl 5):1–270

Results: 696 pts were treated; 58% received all 6 Ra-223 injections. At BL: median age was 72 years; 88% of pts were ECOG PS 0–1; pain was reported as: no pain, mild-moderate, and severe in 21%, 52%, and 27% respectively. 60% of pts received prior therapy with docetaxel. For pts treated with concomitant therapy: 22% were with abiraterone; 20% with denosumab; 18% with bisphosphonates and 4% with enzalutamide. Grade 3/4 AEs were reported in 38% of pts; 21% discontinued Ra-223 due to AEs. At the time of analysis median OS was 16 mos [13-not estimated (NE)]. Median time to first SSE was 18 mos [17-NE]; 24% of pts had ≥50% confirmed ALP decrease from BL; 8% had >50% confirmed PSA decrease from BL. In post hoc analyses OS was statistically significantly longer in pts with BL: ALP < 220 U/L vs ≥220 U/L; ECOG PS 0–1 vs ≥2; no pain vs mild-moderate vs severe; concomitant denosumab; concomitant abiraterone. Conclusions: In Ra-223 treated pts, OS appeared to be better in those treated concomitantly with denosumab or abiraterone. Significantly longer OS was observed in pts with a good ECOG PS, no pain and low ALP. Disclosure: No conflict of interest disclosed. V880

3-year safety follow-up of Radium-223 Dichloride (Ra-223) in patients (Pts) with Castration-Resistant Prostate Cancer (CRPC) and symptomatic bone metastases (Mets) from ALSYMPCA Kramer M.W.1, Kuczyk M.A.1, Parker C.2, Vogelzang N.3, Sartor O.4, Coleman R.E.5, Fang F.6, Skjorestad I.7, Nilsson S.8 Hannover Medical School, Urology and Urologic Oncology, Hannover, Germany, 2The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Sutton, United Kingdom, 3Comprehensive Cancer Centers of Nevada, Las Vegas, United States, 4Tulane Cancer Center, New Orleans, United States, 5Weston Park Hospital, Sheffield, United Kingdom, 6Bayer HealthCare, Whippany, United States, 7Bayer AS, Oslo, Norway, 8Karolinska University Hospital, Stockholm, Norway 1

Background: In ALSYMPCA, the first-in-class α-emitter Ra-223 had a highly favorable safety profile and was well tolerated. Safety monitoring of Ra-223 is essential for a complete safety profile. Here are final safety data including long-term follow-up safety 3 years after last pt’s first injection (inj). Methods: Pts received 6 inj and entered designated follow-up from 4 wks after their last inj to 3 years after first inj. Pts were to be evaluated during tx period and 9 follow-up visits. All adverse events (AEs) were collected until 12 wks after last inj; thereafter, only AEs deemed tx-related were collected. Additional long-term safety data were assessed by specific diseases including acute myelogenous leukemia (AML), myelodysplastic syndrome (MDS), aplastic anemia, and new primary cancer in bone or other organs. Results: Safety population (pts receiving ≥1 inj) included 901 pts (Ra-223, n = 600; placebo [pbo], n = 301); 572 pts (Ra-223, n = 405; pbo, n = 167) entered follow-up. 60 pts (Ra-223, n = 49; pbo, n = 11) completed all follow-up visits. Overall, 564 (94%) Ra-223 and 292 (97%) pbo pts had ≥ 1 tx-emergent AE. During long-term follow-up, there were no reports of AML, MDS, or new primary bone cancer. New primary cancers in other organs were identified: 2 Ra-223 (1 bladder, 1 lymph node mets), 3 pbo (2 skin, 1 adenocarcinoma rectum and sigmoideum), and 2 pbo cross-over pts (1 skin, 1 meningioma). Aplastic anemia was reported in 1 Ra-223 pt. Conclusions: Ra-223 remained safe and well tolerated 3 years after the last pt’s first inj. No major safety issues were identified during the ALSYMPCA 3-year long-term follow-up. Disclosure: No conflict of interest disclosed.

Abstracts


CONTENTS AUTHOR INDEX

V881

V882

Enzalutamide in castration-resistant prostate cancer patients progressing after docetaxel – Retrospective analysis of the Swiss Enzalutamide named patient program

Further characterization of the effects on sequential treatment of docetaxel before or after radium-223 dichloride therapy in Castration-Resistant Prostate Cancer (CRPC) patients with symptomatic bone metastases included in the phase 3 ALSYMPCA trial

Papazoglou D.1, Wannesson L.2, Berthold D.3, Cathomas R.4, Gillessen S.5, Rothermundt C.5, Hasler L.6, Winterhalder R.7, Barth A.8, Mingrone W.9, Uhlmann-Nussbaum C.9, von Rohr L.10, von Burg P.11, Schmid M.12, Richner J.13, Baumann S.14, Kühne R.15, Stenner F.1, Rothschild S.I.1 Universitätsspital Basel, Medizinische Onkologie, Basel, Switzerland, 2Istituto Oncologico della Svizzera Italiana, Bellinzona, Switzerland, 3Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland, 4Kantonsspital Graubünden, Chur, Switzerland, 5Kantonsspital, St. Gallen, Switzerland, 6Onkologiepraxis, Biel, Switzerland, 7Kantonsspital, Luzern, Switzerland, 8Bürgerspital, Solothurn, Switzerland, 9Kantonsspital, Olten, Switzerland, 10Privatklinik Linde, Biel, Switzerland, 11Kantonsspital Aarau AG, Aarau, Switzerland, 12Triemlispital, Zürich, Switzerland, 13Lindenhofgruppe, Bern, Switzerland, 14Onkologiepraxis, Winterthur, Switzerland, 15Waidspital, Zürich, Switzerland 1

Introduction: Five new life-extending therapies (abiraterone, radium-223, cabazitaxel, enzalutamide and sipuleucel-T) have recently been approved for the treatment of metastatic castration-resistent prostate cancer (mCRPC), either before and/or after chemotherapy with docetaxel. So far, there is limited evidence for the optimal sequencing of these agents. Methods: Based on the positive results of the AFFIRM trial a named patient program (NPP) for enzalutamide was initiated in Switzerland. Between 08/2012 and 03/2013 a total of 65 patients were included in the NPP. We reviewed clinical records of these patients. Results: Records from 44 patients were accessible. Median age of the population was 76 years (range, 54–84). At the time of data cut-off, 10 patients were still alive. Median overall survival (OS) from diagnosis of mCRPC was 35 months (95% CI 28–41 months). Average number of therapies for mCRPC was 4.8 (range, 2–8). 37 patients (84%) had four or more lines of therapy. Enzalutamide was used as a second, third, fourth, fifth, sixth or seventh-line therapy in 13%, 20%, 31%, 20%, 11% and 2%. 42 patients (96%) were previously treated with docetaxel (median number of cycles: 6; interquartile range, 5–8.8) and 36 patients (82%) were pretreated with abiraterone acetate (median duration of treatment: 6 months; interquartile range, 4.8–10.0). Median time of treatment with enzalutamide was 3 months (interquartile range, 2–8). Enzalutamide dose was reduced in 8 patients (18%). 18 patients (41%) showed a PSA decline of >30% and 11 patients (25%) a PSA decline of >50% on enzalutamide. Median OS from beginning of enzalutamide therapy was 6.8 months (95% CI 4.1–9.7). The radiographic objective response rate (ORR) for enzalutamide treatment was 6.8% (3 patients) with a median PSA progression-free survival (PFS) of 3.0 months (95% CI 1.6–4.4). 22 patients (50%) received at least one further line of therapy. Data for different sequences will be presented at the meeting. The limitations of the study are mainly due to its retrospective design and the small number of patients treated with some of the sequences. Conclusions: Enzalutamide is an active agent in men with previously treated mCRPC with more than 40% of patients showing a PSA response. In this pretreated population enzalutamide led to median PFS of 3.0 months and a median OS of 6.8 months. The optimal sequencing of novel therapies for mCRPC has to be defined further in clinical trials. Disclosure: Dimitrios Papazoglou: No conflict of interest disclosed. Sacha Rothschild: Financing of Scientific Research: Für Advisory Boards von Astellas, Bayer und Sanofi-Aventis.; Expert Testimony: Astellas, Sanofi-Aventis.; Other Financial Relationships: Honorare für eingeladene Referate von Astellas.

Wedel S.A.1, Mellado B.2, Sartor O.3, Vogelzang N.4, Hoskin P.5, Nilsson S.6, Coleman R.7, Parker C.8, Wahba M.9, Haugen I.10, Shore N.11 Ortenau Klinikum Offenburg-Gengenbach, Urologie und Kinderurologie, Offenburg, Germany, 2Hospital Clinic, Dept. of Medical Oncology, Barcelona, Spain, 3Tulane Cancer Center, Depts. of Medicine and Urology, New Orleans, United States, 4Comprehensive Cancer Centers of Nevada, Dept. of Medical Oncology, Las Vegas, United States, 5Mount Vernon Hospital Cancer Centre, Dept. of Clinical Oncology, Northwood, United Kingdom, 6Karolinska University Hospital, Dept. of Oncology, Stockholm, Sweden, 7Weston Park Hospital, Dept of Oncology, Sheffield, United Kingdom, 8The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Dept. of Clinical Oncology, Sutton, United Kingdom, 9Bayer HealthCare, Whippany, United States, 10Algeta ASA (Bayer), Dept. of Clinical Research, Oslo, Norway, 11Carolina Urologic Research Center, Dept. of Urologic Oncology, Myrtle Beach, United States 1

Introduction & Objectives: Among 921 randomized pts in ALSYMPCA, 526 (57%) had prior docetaxel (D+) and 395 (43%) had no prior docetaxel (D−). Pt characteristics and safety for docetaxel (D) subgroups are presented. Material and methods: ALSYMPCA pts had progressive, symptomatic CRPC with ≥2 bone mets, had no known visceral mets, and had D+ or were unfit for D, declined D, or D was unavailable(D−). OS and SSE data were analyzed using a log-rank test. A post hoc safety analysis of pts who received chemotherapy after Ra-223 (n = 93) or pbo (n = 54) was performed. Results: Baseline characteristics were similar between subgroups.Median OS was significantly prolonged with Ra-223 versus pbo, regardless of D use (D+, HR = 0.70; D−, H = 0.69). Ra-223 reduced risk of SSEs versus pbo, regardless of D use (D+, HR = 0.62; D−, HR = 0.74). Frequencies of grade 3 or 4 hematologic and nonhematologic adverse events (AEs) were low. Among Ra-223 pts, D− pts, versus D+ pts, had lower rates of grade 3 or 4 anemia (11% vs 14%), neutropenia (1% vs 3%), and thrombocytopenia (3% vs 9%). In a post hoc analysis of 147 pts who received chemotherapy after studydrug (of whom 66/93 Ra-223 and 39/54 pbo pts received D), no major safety concerns were identified and incidences of grade 3 or 4 anemia and neutropenia were similar between Ra-223 and pbo pts; 3 Ra-223 pts had thrombocytopenia. Conclusions: Ra-223 significantly prolonged OS with a favorable safety profile in CRPC pts with symptomatic bone mets, regardless of D use. D+ pts had higher rates of grade 3 or 4 hematologic AEs with Ra-223. The incidence of selected hematologic AEs remained low in pts receiving chemotherapy after Ra-223. Ra-223 is an option for pts with CRPC and symptomatic bone mets, regardless of prior D use. Disclosure: Steffen Wedel: Advisory Role: Fa Bayer, Fa Algeta ASA; Financing of Scientific Research: Dokumentationsentgelte Studienteilnehmer; Other Financial Relationships: Teinahme an klinischen Studien. Neal Shore: Employment or Leadership Position: Fa Algeta ASA; Advisory Role: Fa Algeta ASA, Fa Bayer; Financing of Scientific Research: Gehalt Algeta ASA. V883

High-intensity focused ultrasound (HIFU) of prostate cancer – first clincal results with Focal One® Kuru T.H.1, van Essen J.1, Schrading S.2, Bruhn R.2, Heidenreich A.1 RWTH Aachen, Klinik für Urologie, Aachen, Germany, 2RWTH Aachen, Klinik für Radiologie, Aachen, Germany 1

High-intensity focused ultrasound (HIFU) is a technology to ablate prostate cancer. The next generation Focal One® device has been used at various prostate cancer centers in Europe since 2014. It displays substantial improvements: fusion of multiparametric MRI (mpMRI) and transrectal ultrasound (TRUS) images, doubled lesion height enabling treatment of

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

267


CONTENTS AUTHOR INDEX

larger glands, dynamic focusing and intraoperative effectiveness monitoring by contrast-enhanced ultrasound. Precise treatment area contouring within the gland for focal therapy has now become easier. This study describes results obtained in patients treated with Focal One® at the RWTH Aachen university. 14 patients were treated with Focal One®: 7 patients recieved focal or partial ablation and 7 patients recieved whole glande treatment. 9 patients had primary prostate cancer treatment with Focal One® and 5 patients recieved salvage therapy (4 IMRT, 1 low dose brachytherapy). 12 patients had a multiparametric MRI prior treatment. The initial Gleason Score was 7 in median, the mean PSA 9,1 ng/ml. The mean hospital stay was 2,4 days, mean transurethral catherazation time 1,4 days. One patient had to undergo transurethral resection of the prostate 2 weeks after treatment due to high volume of residual urine. No other major complications are noticed until today. In conclusion, the new procedure has a remarkably low morbidity compared to previous generations of devices. For an assessment of oncological outcome and possible late complications the follow-up is still too short. Disclosure: No conflict of interest disclosed.

Freier Vortrag

Lungenkarzinom II V884

Comprehensive multiplex genotyping in lung cancer: challenges and opportunities of interdisciplinary networking using the example of the Network Genomic Medicine Kostenko A.1, Kron F.1, Scheffler M.1, Michels S.1, Sueptitz J.1, Fischer R.1, Merkelbach-Bruse S.2, Scheel A.2, De-Mary P.3, Glossmann J.-P.1, Buettner R.2, Wolf J.1, Netzwerk Genomische Medizin Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany, 2Uniklinik Köln, Institut für Pathologie, Köln, Germany, 3AOK Rheinland/Hamburg, Düsseldorf, Germany 1

Introduction: Targeted treatment of moleculary defined lung cancer subgroups like EGFR mutated or ALK positive lung cancer has become standard now for advanced lung cancer patients based on its superiority over chemotherapy. Still, broad clinical implementation of molecular diagnostics and personalized cancer care is hampered in most European countries by insufficient molecular screening, missing cost reimbursement for comprehensive genotyping and lack of access to appropriate drugs. The German Network Genomic Medicine (NGM) Lung Cancer is a health care provider network now offering next generation sequencing (NGS)-based multiplex genotyping on a central diagnostics platform for all inoperable lung cancer patients in Germany. Methods: The NGS marker panel used for NGM patients consists of 14 genes and 102 amplicons to cover potentially targetable aberrations and is updated each 3 months. NGS evaluation includes frequency of testing, assessment of the time period needed for molecular analyses, accuracy of diagnostics by comparison with alternative methods, assessment of the frequency of personalized cancer therapies triggered by molecular diagnostics, survival in collaboration with established cancer registries and cost-efficacy. Results: Since 2010, when NGM was initiated, the number of participating centers has increased continuously and reached 220 in Q1 2015. In parallel the number of patients annually genotyped within NGM has increased to 5000 in 2014 representing about 10% of the German lung cancer patients. Within NGM for all targetable mutations clinical trials, predominantly phase I-II, could be initiated and enabled us to offer personalized treatment also for mutations without approved drugs (e.g. genetic aberrations in RET, ROS1, T790M and others). In 2014 NGM and the AOK Rheinland/Hamburg, one of the largest German sickness funds, have established the first flat rate cost reimbursement model for NGSbased comprehensive lung cancer genotyping in Europe.

268

Oncol Res Treat 2015;38(suppl 5):1–270

Conclusions: NGM has achieved successful implementation of personalized cancer therapy into clinical routine in Germany including a centralized, quality-controlled multiplex testing platform based on NGS-technology, the integration of clinical partners with different levels of specialization, counseling of partners with regard to therapeutic decisions, establishment of an early-proof of concept trial platform, development of a cost-reimbursement model and joint evaluation programs. Disclosure: Anna Kostenko: No conflict of interest disclosed. Juergen Wolf: Expert Testimony: Novartis, Pfizer, AstraZeneka, BI, Roche. V885

KEAP1-mutations and NFE2L2-mutations in patients with non-small cell lung cancer (NSCLC) Frank R.1, Scheffler M.1, Michels S.1, Eisert A.1, Gogl L.1, Fischer R.1, König K.2, Merkelbach-Bruse S.2, Serke M.3, Ko Y.-D.4, Gerigk U.5, Geist T.6, Heukamp L.2, Büttner R.2, Wolf J.1 Uniklinik Köln, Klinik 1 für Innere Medizin, LCGC, Köln, Germany, 2Uniklinik Köln, Institut für Pathologie, Köln, Germany, 3Lungenklinik, Hemer, Germany, 4 Johanniter Krankenhaus, Bonn, Germany, 5Evangelische Kliniken Johanniterund Waldkrankenhaus, Bonn, Germany, 6Klinik für Pneumologie, Allergologie, Schlaf- und Beatmungsmedizin, Kaiserswerther Diakonie, Düsseldorf, Germany 1

Background: Mutations in genes of the KEAP1-NFE2L2 pathway in patients with NSCLC are associated with an increased tumor growth, resistance towards cytostatic drugs and reduced survival rates. KEAP1 suppresses NFE2L2 under physiological conditions. Oxidative stress or electrophiles cause NFE2L2 to stabilize and translocate to the nucleus, resulting in transcription of various cytoprotective genes. Mutations in KEAP1 and NFE2L2 are described for diverse tumor entities and often cause an increased level of NFE2L2 leading to resistance of cancer cells against anti-cancer drugs and irradiation. This study was performed to characterize KEAP1-mutated and NFE2L2-mutated NSCLC clinically and genetically. Patients and methods: Tumor tissue collected from 446 patients within a regional screening network was analysed for KEAP1 mutations and NFE2L2 mutations using next-generation sequencing (NGS). Clinical, pathological and genetic characteristics of these patients are described and compared with a control group of patients without KEAP1 mutation and without NFE2L2 mutation. Results: We identified 33 patients with KEAP1 mutations. Among these we found 34 different mutations, of which the majority was not previously described. KEAP1 mutations were not restricted to a special exon. In 30 patients (90.9%) additional driver aberrations could be detected. KEAP1 mutations occurred in both genders (male/female ratio 3/1), in squamous-cell carcinoma (36.4%) and adenocarcinoma (60.6%) and were significantly associated with smoking. We also identified 26 patients with NFE2L2 mutations. Among these we found 15 different mutations, of which W24R and E79K were the most common. In 20 patients (76.9%) additional driver aberrations were found. NFE2L2 mutations occurred in squamous-cell carcinoma (69.2%) and adenocarcinoma (23.1%) and were significantly associated with smoking as well. NFE2L2 mutations also occurred in both genders with 61.5% male and 38.5% female. Two patients had both a KEAP1 mutation and a NFE2L2 mutation. Conclusions: Our data suggest a role of KEAP1-mutations and NFE2L2-mutations as a cofactor in addition to classical driver mutations underlying the malignant phenotype of lung cancer cells. So far, this is the largest cohort of patients with KEAP1-mutations and NFE2L2-mutations analysed and described. Further survival and treatment analyses will reveal the role of these mutations for the outcome of these patients. Disclosure: Rieke Frank: No conflict of interest disclosed. Jürgen Wolf: Advisory Role: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Financing of Scientific Research: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Expert Testimony: Bayer, Boehringer-Ingelheim, Novartis, Pfizer, Roche.

Abstracts


CONTENTS AUTHOR INDEX

V886

V887

EGFR mutation testing and treatment with tyrosin-kinase inhibitors in patients with metastatic non-small cell lung cancer treated by office based medical oncologists in Germany – data from the clinical registry on lung cancer (TLK)

Clinical and molecular characteristics of non-small cell lung cancer in patients harboring CTNNB1 mutations

Bertram M.1, Petersen V.2, Heßling J.3, Tessen H.W.4, Münz M.5, Jänicke M.5, Spring L.5, Marschner N.6 Hämatologisch-Onkologischer Schwerpunkt, Hamburg, Germany, 2Praxis Dr. med. Volker Petersen, Heidenheim, Germany, 3Schwerpunktpraxis HämatologieOnkologie, Berlin, Germany, 4Onkologische Schwerpunktpraxis, Goslar, Germany, 5iOMEDICO, Freiburg, Germany, 6Praxis für interdisziplinäre Onkologie & Hämatologie, Freiburg, Germany 1

Introduction: Biomarkerprofiling and individual targeted therapies are the focus of current lung cancer research. EGFR mutations are now well described, and since 2005, tyrosine-kinase inhibitors (TKIs) have been improved for the targeted treatment of patients with specific subtypes of EGFR mutation. Here, data on EGFR mutation testing and TKI treatment in different subgroups of patients with NSCLC are presented. Methods: From 2010 to 2013, 110 sites recruited 2509 patients into the clinical registry on lung cancer. Data on patient and tumor characteristics, biomarker testing and routine treatment administered by office based medical oncologists are documented. The registry also prospectively collects data on outcome parameters, e.g. real-life progression free and overall survival. Patients are followed until death or up to three years. 1400 patients eligible for analysis received palliative treatment for non-small cell lung cancer (NSCLC) and were recruited at the start of 1st-line therapy. Results: Patients were at mean 66 years at start of 1st-line treatment, 63% had adenocarcinoma, 69% were male, 13% were non-smoker, and for 14% KRAS mutation status was tested. Logistic regression revealed the following characteristics to correlate with EGFR testing (Odds ratio > 1, p < 0.05): adenocarcinoma, female gender, non-smoker, KRAS test performed. Overall, 42% (592/1400) of patients with NSCLC and 56% (489/871) of patients with adenocarcinoma were tested for EGFR mutation. Testing of patients with adenocarcinoma has increased from 49% (93/191) in 2010 to 65% (137/212) in 2013. Overall, 17% (98/592) of tested patients with NSCLC and 19% (91/489) of tested patients with adenocarcinoma carried EGFR mutations. Mutations were most frequent in adenocarcinoma of women (24% (47/194) vs. 15% (44/295) in adenocarcinoma of men) and adenocarcinoma of non-smokers (38% (31/82) vs. 19% (13/67) in patients who quit smoking over 10 years ago and 14% (13/90) in smokers). In total, 69% (68/98) of patients with NSCLC and EGFR mutation received TKI within palliative treatment. 1st-line treatment with TKI of patients with EGFR mutated adenocarcinoma increased from 48% (10/21) in 2010 to 67% (14/21) in 2013. Conclusions: Today EGFR mutation status is tested in the majority of patients with NSCLC adenocarcinoma by office-based medical oncologists in Germany. The majority of patients carrying EGFR mutation also receive TKI. Both, testing frequency and TKI treatment are still increasing. Disclosure: No conflict of interest disclosed.

Gogl L.1, Scheffler M.1, Ihle M.2, Michels S.1, Fischer R.1, Serke M.3, Gerigk U.4, Wömpner C.1, Krüger S.5, Kaminsky B.6, Schulte W.7, Höffken G.8, Merkelbach-Bruse S.2, Büttner R.2, Wolf J.1, Lung Cancer Group Cologne University Hospital Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Köln, Germany, 2University Hospital Cologne, Institute of Pathology, Köln, Germany, 3Lungenklinik Hemer, Hemer, Germany, 4Evangelische Kliniken Johanniter-und Waldkrankenhaus Bonn, Bonn, Germany, 5Florence-Nightingale Hospital, Düsseldorf, Germany, 6 Krankenhaus Bethanien, Solingen, Germany, 7Malteser Hospital Bonn, Bonn, Germany, 8Hospital Coswig, Coswig, Germany 1

Background: Although somatic mutations of CTNNB1 in lung cancer have been described, there is still lack of information about prevalence, genetic variability, occurrence of additional aberrations and influence on outcome. This study was performed to analyze CTNNB1 mutations in NSCLC genetically and clinically. Patients and methods: Tumor tissue collected from 3885 patients within a regional screening network was analyzed for CTNNB1 mutations using next generation sequencing (NGS). Clinical, pathological and genetic characteristics of these patients are described and compared with a control group of patients without CTNNB1 mutation. Results: We have identified 58 (1.5%) CTNNB1-mutated patients, whereof 51 have been analyzed so far. This cohort consisted of 32 female and 19 male patients. Adenocarcinoma histology was found in 42 patients (82.4%), but CTNNB1 mutations were also found in squamous cell and neuroendocrine carcinomas. 21 different CTNNB1 mutations were detected on exon 3, of which most are miss-sense mutations (49) besides 2 deletions. The most frequent mutations were S37F and S37C substitutions which each occurred in nine patients. Exclusive CTNNB1 mutations were only detected in five patients. In all remaining patients an additional driver mutation was found including mutations in KRAS, EGFR, BRAF, AKT, PIK3CA and ERBB2 as well as MET amplification, RET-KIF5b-Inversion, ROS1-fusion and mutations and polymorphisms of TP53. Conclusion: CTNNB1 mutations occur alone or in combination with other known oncogenic aberrations in NSCLC. Results of the ongoing clinical characterization of the patients as well as the prognostic and predictive impact of CTNNB1 mutations will be presented. Disclosure: Leonie Gogl: No conflict of interest disclosed. Jürgen Wolf: Advisory Role: lecture and consulting fees from AstraZeneca, ­Bristol-Myers Squibb, Boehringer-Ingelheim, Clovis, MSD, Novartis, Pfizer and Roche; Expert Testimony: research support from Bayer, Boehringer-Ingelheim, Novartis, Pfizer, and Roche. V888

Comparison of fluorescence in-situ hybridization and hybrid capture based NGS sequencing for fusion detection in primary tumor specimen of the lung Griesinger F.1, Cappuzzo F.2, Dziadziuszko R.3, Reck M.4, Lantuejoul S.5, Eberhardt W.6, Tiemann M.7, Heukamp L.8, Menon R.8, Heuckmann J.8 Pius-Hospital, Oldenburg, Germany, 2Ospedale Civile di Livorno, Livorno, Italy, Medical University of Gdansk, Gdansk, Poland, 4LungenClinic, Großhansdorf, Germany, 5CHU Albert Michallon, Grenoble, France, 6Uniklinik, Essen, Germany, 7 Hämatopathologie, Hamburg, Germany, 8NEO New Oncology AG, Köln, Germany 1 3

Introduction: In recent years, the search for novel driver oncogenes has unraveled several gene fusions driving tumor formation in the lung. Here, especially adenocarcinomas harboring ALK and ROS1 gene fusions show striking sensitivity to ALK and ROS1 kinase inhibitors in the clinic, respectively. Thus, molecular diagnostics are an important building block to allow optimal treatment of those patients harboring these gene fusions. The most frequently used technologies to detect gene fusions include fluorescent in situ hybridization (FISH), currently considered as the “gold

Abstracts

Oncol Res Treat 2015;38(suppl 5):1–270

269


CONTENTS AUTHOR INDEX

standard”, immunohistochemistry (IHC), RT-PCR based approaches as well as hybrid capture based NGS sequencing. Methods: Here, we present a selection of primary tumor samples showing discrepant results between fluorescent in situ hybridization and hybrid capture based NGS sequencing. These included samples with positive FISH but negative NEOplus as well as negative FISH and positive NEOplus results. To validate conflicting results, we used available response data to targeted therapies to assess the actual genetic phenotype of the tumor. Results: Overall, several lung adenocarcinomas showed conflicting results between FISH and NEOplus analysis. First, 11 samples were tested positive for ALK rearrangement using FISH which was not confirmed using NEOplus. In line with this finding, one of these tumors did not respond to ALK TKI treatment whereas the other 10 samples showed co-occurring activating EGFR hot-spot mutations. Second, 4 cases were considered as fusion negative by FISH analysis, but were positive for ALK gene fusions when tested with the NEOplus assay. Three out of 4 of these cases showed clinical response to ALK kinase inhibition, while clinical response data for case number 4 is not yet available. In line with the results of the NEOplus assay, one of the responding tumors was also negative for ALK expression using IHC. Conclusions: In summary, we describe a cohort of tumor samples showing discrepant FISH and NEOplus results after testing for gene fusions. However, whenever clinical response data was available, tumor sensitivity was in line with the results obtained by the NEOplus assay. Disclosure: Frank Griesinger: Advisory Role: Pfizer, Roche, Astra Zeneca, New Oncology, BMS, MSD, Novartis, Clovis, Merck, Lilly, Amgen; Financing of Scientific Research: Pfizer, Roche, Astra Zeneca, New Oncology, BMS, MSD, Novartis, Clovis, Merck, Lilly, Amgen; Expert Testimony: Astra Zeneca. Johannes Heuckmann: Employment or Leadership Position: NEO New Oncology AG; Stock Ownership: NEO New Oncology AG.

V889

Experience from the ASCEND-1 trial: Ceritinib in patients (Pts) with ALK-rearranged (ALK+) Non-Small Cell Lung Cancer (NSCLC) and brain metastases Thomas M.1, Schuler M.2, Potzner M.3, Szczudlo T.4, Sutradhar S.4, Yovine A.5, Wolf J.6 Thoraxklinik Heidelberg, Abteilung Onkologie Innere Medizin, Heidelberg, Germany, 2Uniklinik Essen, Innere Klinik (Tumorforschung), Essen, Germany, 3 Novartis Pharma GmbH, Nürnberg, Germany, 4Novartis Pharmaceuticals Corporation, East Hanover, United States, 5Novartis Pharma AG, Basel, Switzerland, 6Uniklinik Köln, Klinik I für Innere Medizin, Köln, Germany 1

Introduction: A common challenging cause of disease progression in ALK+ NSCLC pts including those receiving crizotinib (CRZ) are brain metastases (BM). The oral ALK inhibitor (ALKi) ceritinib showed a 20-fold greater potency than CRZ in enzymatic assays and blood-brain barrier penetration in preclinical studies. Furthermore, in the pivotal ASCEND-1 trial, ceritinib showed intracranial activity. Here, further efficacy and safety were evaluated in pts with BM from ASCEND-1. Methods: ALK+ NSCLC pts, including those with clinically/neurologically stable BM, received ceritinib at a dose of 750 mg/day. Pts in the April 2014 data cut-off with baseline and post-baseline CT/MRI scans were retrospectively analyzed by 2 independent neuro-radiologists which were blinded to systemic response and investigator assessment. Results: Overall, 246 pts were included in the study; 83 were ALKi naïve and 163 ALKi-pretreated. Overall whole-body response rate for ALKinaïve and ALKi-pretreated pts was 72.3% (95% Confidence Interval [CI]: 61.4, 81.6) and 56.4% (95% CI: 48.5, 64.2); progression-free survival was 18.4 (95% CI: 11.1, NE) months and 6.9 (95% CI: 5.6, 8.7) months, respectively.Of 94 pts with measurable or non-measurable BM, intracranial disease control rate (complete and partial response [PR] + stable disease) for ALKi-naïve (n = 19) and ALKi-pretreated (n = 75) pts was 78.9% (95% CI: 54.4, 93.9) and 65.3% (95% CI: 53.5, 76.0). Median ceritinib treatment exposure was 49.6 weeks (range: 7.9–83.0) and 40.6 weeks (range: 0.4–95.1). Six of 11 pts with measurable (RECIST v1.1) BM who had not received prior radiotherapy achieved PR. Among 22 (out of 94) pts with CR/PR, median time to intracranial response was 6.1 weeks. Most common adverse events (all grades) in all pts and the BM subset were diarrhea (86.6% vs 81.9%), nausea (83.3% vs 86.2%), and vomiting (61.0% vs 66.0%). Conclusions: Ceritinib shows clinically significant and durable efficacy in ALK+ NSCLC pts including pts with BM. Disclosure: Michael Thomas: Advisory Role: Astrazeneca, Novartis, Lilly, Roche, Pierre Fabre, MSD, BMS; Financing of Scientific Research: Astrazeneca, Novartis, Lilly, Roche, Pierre Fabre, MSD, BMS. Jürgen Wolf: Advisory Role: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Financing of Scientific Research: AstraZeneca, BMS, Boehringer-Ingelheim, Clovis, Novartis, Pfizer, Roche; Expert Testimony: Bayer, Boehringer-Ingelheim, Novartis, Pfizer, Roche.

270

Oncol Res Treat 2015;38(suppl 5):1–270

Abstracts


CONTENTS AUTHOR INDEX

Author Index Oncol Res Treat 2015;38(suppl 5):272–288

A Abdel-Wahab O.  V420 Abdollahi A.  V583 Abendroth A.  V25, V27 Abenhardt W.  V75 Aberer F.  P443 Aberger F.  V664 Abhari B.A.  V299 Abovyan M.  P225 Abramczyk M.  V25, V27 Abruzzese E.  V604 Achenbach H.  V721 Acton G.  V424 Adamek J.  V635 Adamova Z.  P811 Adenis A.  P211 Advani R.H.  V73 Adzersen K.-H.  V569 Agrawal M.  V538 Ahci M.  V849 Aigner M.  P816 Ajib S.  V161 Akca A.  P494 Akmut F.  P184 Aksnes A.  P264 Al-Ali H.K.  P173, P187, V725 Alashkar F.  P479, V72 Alawi M.  V390, V627 Al-Batran S.-E.  P214, P262, P827 Albers P.  P271, V108 Albert M.  V127 Aldaoud A.  P783, P827, V26 Alder J.  P488 Aldrian C.  V551 Alexis M.  V285 Alghisi E.  V342, V582 Al-Matary Y.S.  V33, V632 Alsdorf W.  P269, V878 Alt-Epping B.  P801, V733 Altmann B.  V299 Altmann T.  P175, V131, V134 Amann A.  P500 Amann E.  V130 Amann K.  V878 Ambrosetti A.  V584 Amram M.-L.  P263 Amtmann R.  P248 Andel J.  P453 Anderson K.C.  V548 Andre M.  V850 Andrea M.  V43 Andrews D.  V309 Andrulis M.A.  V420 Angenendt L.  V717 Anhuf J.  P458 Anna S.  V164 Annolleck T.  V666 Ansén S.  V44 Anthoney A.  P824

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

Antunes E.  V130 Anz D.  V24 Apostolou P.  P825 Apostolova P.  V161 Appel N.  P202 Apperley J.F.  V604 Arn M.  V431 Arnd J.  P221 Arnold R.  P469, V161, V317, V859 Arnold S.  V616 Arsenic R.  P495 Arteaga M.F.  V717 Asslaber D.  V664 Atanackovic D.  P773 Atzpodien J.  V392 Auer R.  V73 Aul C.  P501, P800, V118, V119 Aulitzky W.E.  V350 Austein T.  P762 Autenrieth S.E.  V636 Avemarg S.  V435 Avlar M.  V583 Ayuk F.  V161

B Baccarani M.  V85, V600, V603, V604 Bach A.  P219 Bacher U.  P781, P782, V119 Bachhuber P.  P757 Bachinger A.  V115 Bachmann H.S.  P259 Bacigalupo A.  V164 Back W.  P762 Backert L.  P472, V550, V686 Bader P.  V860 Baerlocher G.M.  V602 Baher L.  V73 Bahlo J.  V290, V292, V580, V666 Bähring R.  V878 Bainschab A.  P245 Bajrami Saipi M.  P185 Bakthiar S.  V860 Baldus C.D.  P231, V286, V319, V426, V542 Balermpas P.  V583 Balic M.  V597 Balleisen L.  V601 Balló H.  V418, V419 Balser C.  V81, V418, V419 Balsiger C.  P489 Balzarotti M.  V584 Bangerter M.  V722 Bár T.  P811 Barckhausen C.  P203 Bargou R.  P786, V39 Barlow S.  V127 Baron F.  V118 Barone C.  P211 Barrientos J.C.  V73, V293

Bartels M.  V289 Barth A.  V881 Barth J.  P171, V418, V419 Bashari M.H.  V431 Bassermann F.  V39, V299, V547 Bastholt L.  V93, V613 Bastian L.  V286 Bauder-Mißbach H.  V620 Bauer C.K.  V878 Bauer G.  P238 Bauer S.  P804, P808, V553, V579 Baumann M.  V583, P483 Baumann S.  V881 Baumann U.  V299 Baumann W.  P799, V749, V750 Bäumer N.  P202 Bäumer S.  P202 Baumgartner U.  V76 Baumhardt M.  P460 Baur R.  P198 Beaupre D.M.  V73 Becher C.  P236 Beck J.  V285 Beck R.  V744 Becker G.  P258 Becker H.  V118 Becker M.  V350 Becker N.  P506, V569 Becker S.  P189 Beckhove P.  V431 Beckmann G.  P201 Beelen D.W.  V124, V161, V162, V849 Beham-Schmid C.  P218 Behre G.  V126, V428, V541 Behringer D.M.  V46 Beier F.  P232, V300 Beinemann J.  V615 Beissbarth T.  P205 Bek S.  V132 Belka C.  V583 Beller M.  V301, V423 Belleville E.  P458 Ben Batalla I.  P773 Benboubker L.  V548 Bender R.  V556 Benes V.  P186 Benkler T.  V616 Benner A.  V38 Bennett J.M.  V114, V119 Benser J.  P799 Berdel W.E.  P202, P754, V37, V426, V429, V717 Berenstein R.  P770, V542 Berg T.  P171 Berger C.  P447 Berger D.  V621 Berger K.  P240, P517, V745 Berger T.  P488 Bergmann L.  P266, P267


CONTENTS AUTHOR INDEX

Bergmann M.  V292 Bergmann U.  V126 Berkessel A.  V665 Bernd H.-W.  V421 Berning P.  P805, P807 Berthold D.  V881 Bertram M.  V886 Bertsch T.  V685 Bertsch U.  V38, V40 Bertz H.  V161, V742, V746 Besseler M.  V567 Besses C.  V721 Bethge W.A.  P444, V858, V861 Betticher D.  P260 Beurskens F.J.  P793 Beuthien-Baumann B.  P810 Beutner D.  V94 Beykirch M.  V359 Beylich A.  P799 Bezold K.  V569 Bhatti A.  V135 Bianconi D.  P499 Biehl L.M.  V18, V580, V618 Bielack S.  V104 Bigalke I.  V131 Bill M.  V126, V428, V541 Bilstein A.  V691 Binder M.  P220, P773, P781, P782, V390, V584 Birgegard G.  V721 Birnbaumer L.  V351 Bischoff S.  P826 Bisht S.  P829 Bittenbring J.  P191, P192, V300 Bittner A.  P474 Blachke P.  V747 Blagitko-Dorfs N.  V714 Blaise D.  V164 Bläker H.  P826 Blank N.  P243 Blasczyk R.  V709, V711 Blau I.W.  P770, V542 Blau O.  P770, V542 Blau W.  V418, V419 Blay J.-Y.  P803, V579 Blazar B.R.  V161 Bleckmann A.  P205 Bleickardt E.  V548 Bloch W.  P823 Blommer J.  P784 Blum K.A.  V73 Blumstein N.  V555 Bob R.  V80 Boch T.  V625, V626 Bochum S.  P511 Bodden G.  P247 Bodis S.  V324 Body J.-J.  V732 Bogatyreva L.  P179 Bogner C.  V309 Bohle R.-M.  V302, V304, V704 Bohlen J.  P217 Bojko P.  V567

Author Index

Bokemeyer C.  P200, P206, P207, P208, P209, P210, P263, P269, P773, P782, P822, V390, V556, V878 Bollig A.  V745 Bommer M.  P480, P481 Bondong A.  P439, P471 Bonin M.  P184 Boqué C.  V85 Borchmann P.  V350 Borges jr.U.  P799 Bornhäuser M.  V37, V163, V429, V641, V855 Bòrquez D.  P252 Bosmann M.  P819 Bosnar S.  P795 Bosserhoff A.  P816 Botezatu L.  V33, V34, V611, V632, V852 Böttcher S.  V289, V292, V571, V666, V834 Böttger I.  V606 Bouché O.  P211 Bouillon A.S.  P232 Boultwood J.  V852 Božić T.  V345 Braciak T.A.  V133 Bradley-Garelik B.  V85 Braess J.  V685 Brägelmann J.  P246 Braig F.  P781, V390 Brand S.  P439 Brandauer K.  V847 Brandt J.  V863 Branle F.  P465 Brass V.  V752 Braulke F.  V117, V119 Braun M.  P181, V434 Brauneck F.  P175 Brecht P.  P510 Bremm M.  V860 Brendel C.  P203, P273, P765 Brenn J.  P450 Briest F.  P495 Brinckmann F.  V683 Bronisch O.  V359 Brors B.  V687 Brose M.S.  V91, V93, V613 Brossart P.  P180, P246, P254, P257, P482, P758, P760, P813, P829, V82, V425, V552, V723, V848 Broszeit-Luft S.  P473, V394, V395, V682 Brown J.R.  V291 Bruch H.-R.  V115, V682 Bruckner T.  V863 Brudler O.  P757 Brüggemann M.  V13, V131, V287, V289 Brugger W.  V418, V419, V714 Bruhn R.  V883 Brümmendorf T.H.  P232, P754, P761, P760, V426, V605 Brummer T.  V616 Brune M.  V161 Brunkhorst F.M.  V851 Brunner A.  V299 Bruns H.  V300, V552 Bryja V.  V706 Bscheider M.  V132

Buchheidt D.  V625, V626 Buchholz F.  P202 Büchner-Steudel P.  V392 Buck M.  P504 Bücklein V.  P170, P493, P806, V131 Bückner U.  P255 Budach V.  V583 Buehring H.-J.  V850 Buess M.  P467, V310 Buettner R.  V884 Bug G.  V161, V714 Bugl S.  V662 Bulduk M.  V36, V165 Bullinger L.  V32, V35, V39, V538, V559, V716 Bulycheva E.  V429 Bunjes D.  V125, V128, V129 Burchardi N.  V432 Burchardt A.  V418, V419 Burchardt M.  P445 Burchert A.  P470, V161, V601, V605 Burgstaller S.  P453 Burmeister T.  P470 Burnett C.  V344 Busch C.-J.  P822 Busemann C.  P436 Buser A.  V41 Buske C.  V559 Buslei R.  V302 Buss E.C.  V655 Busse A.  P495 Butterfaß-Bahloul T.  V426 Buttkereit U.  V849 Büttner H.  V627 Büttner M.  V300 Büttner R.  P468, V44, V49, V557, V885, V887 Byrgazov K.  P448

C Cadeddu R.-P.  V545, P174 Call T.G.  V291 Camargo V.  V579 Canis M.  V733 Cao B.  V341 Cappuzzo F.  V888 Capraro J.  V66 Cardone M.H.  V431 Carroll K.J.  V582 Casali P.G.  P803 Casanova L.  V85 Caspar C.  P260 Castagna L.  V164 Cathomas R.  P260, P821, V881 Cathomen T.  V386 Cavalli F.  V584 Cavanna D.  P757 Caysa H.  P204 Cayuela J.-M.  V285 Cazorla Arratia P.  P465 Cazzola M.  V114, V852 Cermak J.  V114 Cerny-Reiterer S.  V621 Chakupurakal G.  V166

Oncol Res Treat 2015;38(suppl 5):271–288

273


CONTENTS AUTHOR INDEX

Chatterjee M.  P786 Chawla S.  V579 Chemnitz J.  P784 Chen R.  V579 Chen W.  V660 Cheng M.  V73 Chevallier P.  V164 Chi X.  V309 Chifudov S.  P470 Chillà A.  P499 Chiroli S.  P450 Chmielowska E.  V73 Chmielowski B.  V579 Chott A.  V422 Choy E.  V579 Christ H.  V618 Christ T.  V878 Christensen B.  V432 Christner M.  V627 Christofori G.  V310 Chu A.D.  V291 Chuah C.  V85, V604 Chung J.  V613 Cislo P.  P265 Clackson T.  V600, V604 Claus B.  V626 Claus R.  V714 Claussen A.  P486 Clow F.  V291 Coleman R.E.  V880, V882 Colita A.  V603 Combs S.E.  V583 Conradi I.  P466 Conradi L.-C.  P205 Coon C.  V48 Corbacioglu A.  V32, V538 Corbacioglu S.  V692 Cornely O.A.  P797, P798, V580, V618, V625, V626 Cornez I.  P813 Corradini P.  V164 Cortes J.E.  V83, V84, V85, V424, V600, V604 Coutre S.E.  V291 Craig A.R.  V424 Craig M.D.  V424 Cramer P.  V290, V292, V580, V666 Cremer M.  P228, V659 Crippa S.  V90, V92 Crivello P.  V124, V849 Cross M.  P173, V126, V428, V541 Cross N.C.P.  V581, V724 Crysandt M.  V714 Csomor J.  P475 Cubas Cordova M.  P773 Cwynarski K.  V584

D D'Adamo D.  V579 D'Addio A.  P457 Daecke S.N.  V82 Dahlfrancis M.  P812 Dahmen U.  P828 Damm K.  P513 Damnali G.  P507

274

D'Amore F.  P227 Danhof S.  P772, P778, P787 Däßler K.-U.  V45 Dastani H.  V48 Datta N.  V324 De Benedittis C.  P457 De Cock E.  V732 De la Fouchardiere C.  V93, V613 De Wit M.  P790 De Witte T.  V118 DeAngelo D.J.  V604 Dearden C.  V420 Debus J.  V583 Dechêne A.  V25, V27 Decker T.  P823, V670 Deckert M.  V584 Deininger M.W.  V600, V604 DeKrijger I.  P814 Delfau-Larue M.-H.  V71 De-Mary P.  V884 Demetri G.D.  P803 Dengler F.  V686 Dengler J.  P452, V81, V350 Denkert C.  P826 Denny M.  P794 Depping R.  P817 Derer S.  P793 Derigs H.-G.  P213 DeRosa M.  V48 Derwahl K.-M.  P779 Desax M.C.  V612, V614 Deutsch A.  P218 Dickerhoff R.  V353 Diedrich D.  P796 Dieing A.  P459, P473 Dienst A.  P251 Dierks C.  V616, V617 Dierks S.  V610 Dietlein F.  V557 Dietrich S.  P439, P471, V164, V294, V420 Dietz C.T.  V601, V602, V724 Dirksen U.  P805, P807 Dirsch O.  P828 Distelmaier L.  V353 Ditschkowski M.  V161 Dittmar G.  V660 Dittrich A.  V730 Dittrich T.  V659 Diwok C.  P194 Dobbie M.  P199 Dobrosch L.  P482 Doehn C.  V675 Döhner H.  P480, V32, V125, V129, V292, V425, V538, V559, V620, V716, V722 Döhner K.  V32, V425, V538, V559, V714, V716, V722 Dolak W.  V422 Dölken G.  P436, P455 Dombret H.  V285 Dombrowski-Lütcke M.  P831, V672 Doostkam S.  P224 Dorda A.  P499 Dörfel D.  P444, P812, V688, V744, V861 Dörken B.  P770, V28, V303, V542, V660, V859

Oncol Res Treat 2015;38(suppl 5):272–288

Dornaus S.  P244 Dotterweich J.  V549 Downward J.  P814 Dräger R.  V607 Dreger P.  P439, P471, V125, V164, V294, V863 Dresel I.  V714 Drexler B.  V858 Dreyling M.  P216, P220, P221, P223, P226, V71, V73, V421 Drosten C.  P254 Duecker S.  V656 Dugas M.  V426 Dührsen U.  P473, P479, V33, V34, V72, V353, V611, V632, V852 Dünnebacke J.  V748 Duran Graeff L.  P797 Dürig J.  V40, V420, V632 Dürk H.  V418 Dürkop H.  V80 Düsedau A.  P781 Dutcus C.  V91, V673 Duyster J.  P224, P486, P504, P774, P775, P776, P785, V42, V161, V537, V551, V616, V617, V752, V858 Dyer M.  V420 Dyshlovoy S.  P269, V878 Dziadziuszko R.  V888

E Eberhard N.  P453 Eberhardt W.E.E.  V46, V888, P466 Ebert A.  P487 Ebert R.  V549 Ebner K.  P819 Echchannaoui H.  V130 Ecke T.  P262 Eckoldt J.  V163 Ecsedi M.  V161, V858 Edinger M.  V601, V860 Egerer G.  V38, V74, V625, V626 Egert M.  P255 Egger M.  V392, V714 Eggers H.  P268 Ehninger G.  P483, P496, P754, P810, V37, V163, V426, V429, V611, V641, V852, V855 Eichhorst B.  V290, V292, V580, V666, V833 Eigendorff E.  P244, P447, V605 Eigl S.  V626 Einsele H.  P237, P441, P772, P778, P786, P787, P795, V39, V602 Eisen T.  V673 Eisenschink A.M.  V620 Eisert A.  V49, V885 Eisterer W.  P500, V143, V406, V681 Eiz-Vesper B.  V709 Ekkehard E.  V432 Elisei R.  V91, V93, V613 Elmaagacli A.H.  P438 Elter T.  V420 Enard W.  V343 Enders B.  V306 Endres S.  V24

Author Index


CONTENTS AUTHOR INDEX

Endris V.  P497 Engel E.  V390 Engel K.  V299, V547 Engelhard M.  V421 Engelhardt M.  P486, P504, P774, P775, P776, P785, V39, V42, V96, V551, V618, V752 Engelke A.  V666 Engert A.  P239, V609 Ensinger C.  P500 Epting T.  P504 Erba H.P.  V424 Erck C.  V80 Erdmann-Reusch B.  P491 Erhardt E.  V620 Erle A.  V559 Ernst T.  V619 Esain V.  V582 Eschenburg A.  V301, V423 Escofffre-Barbe M.  V285 Espeli V.  V90, V92 Ettrich T.  V392 Etzrodt M.  V316 Evans T.R.J.  P824 Eveslage M.  V426 Exner A.-K.  V568

F Fabarius A.  P183, P234, P454, V581, V601, V602, V724 Fabbri A.  V584 Fabisch C.  P447, V605 Fabri M.  V300 Facon T.  V548 Fadle N.  V302, V304, V704 Falcone A.  P211 Falge C.  V602 Falk M.  P460, P466, V47 Falkenhorst J.  V553 Fan D.N.-Y.  V660 Fan F.  V431 Fang F.  V880 Färber J.  V628 Farzaliyev F.  P804, P808 Fasching P.  P823 Fätkenheuer G.  V657 Faul C.  P444, V861 Fehm T.  V430 Fehr E.-M.  V35 Fehse B.  P773, V390 Feichtinger J.  P218 Feiten S.  P820, V433, V748 Fekete N.  V559 Feldmann G.  P829 Felipe Fumero E.  V717 Feller A.C.  V421 Fenaux P.  V114 Fend F.  V564 Fenk R.  P174, P247, P251, V753 Fenn N.  V133 Fernandez-Sáiz V.  V299 Ferraro D.  V310 Ferreira M.S.  P232 Ferreri A.J.M.  V584 Feuerbach M.  P510, P515, P518

Author Index

Fey G.H.  V133 Fey S.  P231, P234 Fichtner M.  P220 Fiedler W.  P773, V32, V425, V538 Fiegl M.  P175, P446 Fietz T.  P255, P256, P458, V658 Figueiredo C.  V711 Finel H.  V164 Fink A.-M.  V290, V292, V580, V666 Fink S.  P786 Finke F.  V533 Finke J.  P222, V161, V584, V658, V858 Finkernagel F.  P474 Fisch P.  P440 Fischer I.  V339, V567 Fischer J.C.  V132, V545 Fischer J.R.  V46 Fischer K.  V290, V292, V580, V666 Fischer L.  P215 Fischer M.  V427 Fischer R.  V44, V49, V884, V885, V887 Fischer S.  V555 Fischer T.  P508, V565, V628, V630, V707 Flaherty P.  P829 Flamme H.  V665 Flechtner H.-H.  V565 Fleck E.  P440 Fleischhauer K.  V124, V849 Flöck A.  P180 Florek M.  V344 Flores C.  V848 Florschütz A.  V392 Föll J.  V692 Follo M.  V551, V616 Follows G.  V420 Folprecht G.  P810 Fonatsch C.  V119 Forcher V.  P500 Foreman A.  P451 Forstbauer H.  V432 Förster F.  P823 Fox C.P.  V584 Fox J.A.  V424 Frank M.  V394 Frank O.  P452, P456 Frank R.  V49, V885 Franke G.  V627 Franke G.-N.  P187, P241, V43, V126, V428 Fransecky L.  V286 Frattini M.  V90, V92 Frech M.  P178, P830 Freichel M.  V351 Freigang F.  P510, P515, P518 Freitag A.  P463 Freitag S.  V747 Freund M.  P796 Frey A.  V42 Frey J.  P176 Freysoldt B.  P216 Freystein J.  P204 Frickhofen N.  P189, V426 Friedrich A.  V641 Friedrich S.  P506, V569 Friesenhahn V.  V433, V748 Fritsch A.  V685

Fritsch K.  P814, P222, P224, V658 Fritsch R.  P814 Fritsch S.  V127 Fritz B.  V545 Frobel J.  P756, V345 Fröhler S.  V660 Fröhlich K.  V435, V671 Fröhling S.  P810, V420, V687 Frommer J.  V565, V630 Früh M.  V46, V90, V92, V612, V614 Fulda S.  V299 Füller M.  V426 Füllgrabe M.  V287 Fund N.  P190, P197 Furman R.R.  V291 Fürniß T.  V620 Fürschuß C.  P490 Fürst D.  P469, V128 Fütterer M.  V716

G Gaab J.  P488 Gaehler A.  V420 Gaidzik V.I.  V32, V425, V538 Gaiser T.  V581 Gakis G.  V334 Galceran J.C.  V879 Gallagher M.E.  P517 Gallamini A.  V868 Gambacorti-Passerini C.  V604 Gamperl H.  V689, V690 Ganser A.  P268, V32, V118, V352, V418, V425, V538 Ganser M.  P763, V662 Ganster C.  V610, V854, V856 Garbi N.  P813 Garcia-Manero G.  V114 Garcia-Marquez M.  V166 Garcia-Vargas J.  P261, P264 Garn H.  P474 Gärtner F.  V343 Garz A.-K.  V715, V857 Gassmann W.  P764, V354 Gastl G.  P453, P500 Gatalica Z.  V556 Gatta F.  V732 Gattermann N.  P754 Gauler T.C.  V46 Gautschi O.  V521 Gazawi N.  P255 Gebauer L.  P203 Gebhardt W.H.  V35 Geer T.  P458, P757 Geetha N.  P499 Gehlen H.  V552 Geiger O.  P815, V307 Geiges G.  V554 Geismar D.  P808 Geissler D.  P453 Geissler K.  P453 Geißler M.  V392 Geist T.  V885 Gelderblom H.  V579 Gensch V.  P773

Oncol Res Treat 2015;38(suppl 5):271–288

275


CONTENTS AUTHOR INDEX

Geörg C.  V687 George D.  V579 Georgoula L.  P249 Gerber B.  P796 Gerbitz A.  V300, V552, V691 Gerecke C.  V40 Gerigk U.  V885, V887 Gerken G.  V25, V27, V72 Gerlach C.  P484, V731 Gerloff D.  V541 Germing U.  P174, P232, P233, P247, V114, V116, V118, V119, V606, V644, V714, V753, V852 Gernböck C.  P802 Gerngroß C.  V718 Gerss J.  V426 Gerstner S.  V860 Gerull S.  V41 Geyh S.  P174, V644 Ghia P.  V293 Ghibelli L.  V692 Ghielmini M.  V90, V92, V589 Ghoreschi K.  P198 Giagounidis A.  V118, V119, V426, V714 Giannetta L.  V93 Giannini B.  P457 Giebel B.  V34, V852 Gieseler F.  V159, V689, V690 Gillessen S.  V879, V881 Gillor D.  V657 Girschikofsky M.  V425 Gkika E.  V583 Glaser A.  V427 Gläser D.  P791, V734, V735 Gläser H.  V734, V735 Glaß B.  P240 Gleixner K.  P448 Glen H.  V673 Glimm H.  V420, V687 Glöckner J.  V299, V547 Glossmann J.-P.  V884 Goebeler M.-E.  V601 Goebell P.J.  P266, P267, V674, V675 Goecke T.W.  P756, V345 Goede V.  V290 Gökbuget N.  V285, V286, V287, V289 Gogishvili T.  P772 Gogl L.  V49, V885, V887 Gogoll K.  P794 Göhler T.  P266, P267 Göhring G.  V425, V538, V724 Gökkurt E.  P754 Goldberg R.M.  P211 Goldschmidt H.  P769, V38, V40, V546 Göllner S.  V426 Göner M.  P779 Gooden K.  V83 Goosen R.  V310 Goossens S.  P758 Gorantla S.P.  V537 Görgens A.  V34, V852 Görl N.  P791 Gorlov J.S.  V584 Görner M.  P439, V294, V426 Goßmann J.  P474

276

Göthert J.R.  P754 Göttel R.  V295, V393 Götz M.  V125, V129, V559 Götze K.S.  V32, V119, V343, V425, V426, V544, V714, V715, V718, V853, V857 Goy A.  V73 Grabowski P.  P193, P495 Graeven U.  P468 Graffunder G.  P255 Gralla R.J.  V48 Gramatzki M.  P236, V36, V165, V561 Grassinger J.  V341, V719 Grassmann S.  V24 Gratt J.  V879 Graul K.  P187 Graziani G.  P785 Greenberg P.L.  V114 , V119 Greeve J.  V392 Greger N.  V747 Greiff U.  P800 Greil C.  P219 Greil R.  P453, V31, V664 Greinacher A.  P436 Greiner J.  V125, V129, V559, V605 Greinix H.T.  P245, P437, P443, P489, V399 Grewing V.  V847 Griesinger F.  P460, P462, P464, P466, P468, V47, V888 Griesshammer M.  V425, V527, V721, V722 Grignani G.  V579 Grigoleit G.U.  P237, V161 Grimm J.  V126, V428, V541 Grischke E.-M.  P823, V432 Grishina O.  V42, V714 Grob T.  V390 Grobe N.  P455 Grobholz R.  V369 Gronwald W.  P816 Groschek M.  P783 Gröschel S.  V687 Grosse-Hovest L.  V846, V850 Grossert A.  P488 Große-Thie C.  V734, V735 Groß-Ophoff-Müller, C.  V290, V292 Grosu A.L.  V583 Grothey A.  P201, P211 Grotius K.  V607 Grugel R.  V75 Gruh B.  V427 Grund C.  V533 Grundhoff A.  V390 Grünebach F.  P812, V688 Grünwald V.  P268, P803, V68, V675 Grziwok S.  V343, V543, V544 Gschwend J.E.  P263 Gudzuhn A.  P455 Guger-Halper U.  P248, P485 Gugliotta L.  V721 Guilhot F.  V600, V604 Guillerm G.  V285 Guinney J.  P201 Günter M.  V636 Günther A.  P236, V165 Gurtovaya O.  V293 Gütgemann I.  P482, P758

Oncol Res Treat 2015;38(suppl 5):272–288

Güth U.  V298 Guzii A.  V878

H Haack B.  V426 Haarmann J.  P829 Haas A.  P512 Haas M.  P195, P507, P802 Haas R.  P174, P232, P233, P247, P250, P251, P271, V116, V545, V632, V644, V753 Haas T.  V132 Haase D.  V114, V117, V119, V610, V854, V856 Haaß W.  P454 Habringer S.  V308, V715, V718 Hackanson B.  V714 Hadzijusufovic E.  P755 Haegeman G.  V692 Haeger J.-D.  V671 Haehnel P.S.  V35 Haen S.P.  V686, V744, V850, V861 Haferlach C.  P183, P234, V601, V720, V724 Haferlach T.  P270, V350, V420, V720 Hagemeijer A.  V118 Hähling D.  P455 Hahn J.  V625, V626, V636 Hahn L.  V682 Hahn M.  P471, V294 Hahn-Ast C.  P246, P257 Hähnel P.S.  V306 Haibach M.  V691 Haigh J.  P758 Haj Abdo K.  V555 Halbe L.  P484 Halbsgut N.  V854 Hallas C.  P217, P466, V47 Hallek M.  P784, V166, V290, V291, V292, V420, V580, V665, V666 Halter J.  V41, V161, V858 Haluska F.G.  V600, V604 Hamdani M.  V721 Hamel T.  V620 Hammerer P.  P262 Hammersen J.  P447 Handgretinger R.  V708 Hänel A.  V38 Hänel M.  P754, V40, V426, V601, V605, V656 Hanfstein B.  V602 Hanitsch L.G.  P193, V634 Hansen R.  P754 Hansmann M.-L.  V304, V421, V704 Harde J.  P235, P255, P256, P783, V26, V75 Harenberg M.  P478 Harich H.-D.  P266, P267 Harrison C.  V721 Hart C.  V341, V692, V719 Hartjen A.S.  V165 Hartmann S.  V421, V704 Hartmann T.N.  V664 Hartmann T.  P239 Hartmann U.  P189 Hartwig U.F.  V135, V563 Hasford J.  P458, V601, V602, V603

Author Index


CONTENTS AUTHOR INDEX

Hasibeder A.  P819 Haslbauer F.  P767 Hasler L.  V881 Hass R.  V852 Hatzl S.  V307 Hau P.  V152 Hauber D.  V128 Hauch U.  V81 Hauer A.  V750 Haug U.  P506 Haugen I.  P261, V882 Hauschild J.  P269, V878 Hauschild O.  P774 Häusler C.  P453 Hausmann A.  V127, V164 Healey D.  V84 Hebart H.  P249, V392, V683, V684, V708, V722 Hebestreit N.  V566 Hechmati G.  V732 Hecht A.  P183 Heesch S.  V286 Hegenbart U.  P439, P471, P769, V169 Heger J.  V435, V671 Heger M.  V558 Hegewisch-Becker S.  V26 Hegge N.  P246 Hehlmann R.  P451, V601, V602, V603 Heidegger S.  V132 Heidel F.  V425, V722 Heidemann E.  V683 Heidenreich A.  V879, V883 Heidenreich D.  V285, V862 Heider A.  V418, V419 Heil G.  V418, V714 Heilig C.  V863 Heim D.  V41, V602 Heim M.E.  V567 Heine A.  V82, V723, V848 Heinemann F.M.  V124, V849 Heinevetter B.  V851 Heining C.  V687 Heinke H.  P796 Heinold A.  V124, V849 Heinrich D.  V879 Heinz S.  P488 Heinz W.  V625, V626, P441 Heissner K.  P198, P766 Heitmann J.  V662 Heits F.  P256 Held G.  P191, P192, V32, V656 Held M.  V848 Held S.A.E.  V82 Helle S.  P264 Hellebrand E.  V684 Heller S.  V286 Hellmich M.  V618 Hellmuth J.  V127 Helness A.  V852 Hemmati P.  P489, V317, V859 Hemmaway A.  V584 Henke M.  V752 Henke R.P.  P466 Henkel L.  V343, V544 Henn A.  P271

Author Index

Henne K.  P774, P785 Henrich B.  P250 Henrich F.  P816 Henschler R.  P440 Hensel M.  V655, V657 Hentrich M.  V148, V618, V657 Hentschel L.  P483, P492, P496 Herbst C.  V129 Herfarth K.  V74 Herget G.W.  P774, P785 Herhaus P.  V718 Hering J.  V655 Herling M.  V835 Hermann B.  P503 Hermann S.  P506, V569 Hermes-Moll K.  V749, V750 Hermine O.  V71, V164 Herndlhofer S.  P448 Herold M.  V420 Herold S.  V37 Herold T.  V286 Herr W.  P449, V341, V426, V562, V692, V719 Herrmann A.  V303, V660 Herrmann E.  P266, P267 Herrmann M.  V420 Hertenstein B.  V425 Hertz L.  V351 Herzberg P.Y.  P489 Hess G.  P226, P484, V731 Hess V.  P488 Hesse E.  P773 Hesse J.  P252 Heßling J.  V886 Heuckmann J.  V888 Heukamp L.C.  P259, P468, V885, V888 Heuser M.  V32, V425, V538, V611, V714 Heußel C.P.  V626 Heussner P.  P489, P792 Heymanns J.  P820, V433, V748 Heyn S.  P187, P241, V43 Hiddemann W.  P175, P170, P216, P221, P223, P493, P514, P792, V71, V127, V131, V134, V421 Hieke S.  P775, P776 Hielscher T.  V40 Hildebrandt B.  P233, V116, V119, V682 Hilgendorf I.  P244, P442, P489, V566, V747 Hilger R.A.  V46 Hillengass J.  P769, V38, V546 Hillig G.  P451 Hillmen P.  V291 Hinterleitner C.  P766, P768, P788 Hinterseer E.  V664 Hirschberger J.  P223 Hirt C.  P450, P455, V429 Hitz F.  P177 Ho A.D.  P186, P228, P243, P439, P471, P769, V74, V125, V294, V420, V546, V655, V659, V863 Hoang V.T.  P186 Hochhaus A.  P244, P442, P447, P503, V84, V85, V427, V566, V600, V601, V602, V604, V605, V619, V722, V851 Höchsmann B.  V350

Höckendorf U.  V853 Hoelzer D.  V285 Hoenigl M.  V626 Hofbauer S.  V664 Hofer S.  V744 Höffken G.  V887 Höffkes H.-G.  V432 Hoffmann A.C.  P242 Hoffmann C.  V657 Hoffmann F.  P187 Hoffmann H.  V622 Hoffmann I.  P186 Hoffmann P.  V860 Hoffmeister sen. H.  P493 Hofheinz R.-D.  V26, P213, P214, P262, P827 Höfler S.  V616 Hofmann S.  V125, V129, V559 Hofmann W.-K.  P183, P231, P234, P454, V119, V581, V602, V608, V625, V626, V724, V862, V872 Hoheisel M.  P462, P464 Hohenberger P.  V579 Hohloch K.  V377 Holdenrieder S.  V685 Hollburg W.  P754, P801 Höllein A.  V718 Holler E.  P489, V860 Holtick U.  P784, V161, V166 Holzvogt B.  V43 Holzwarth K.  P480 Homayounfar K.  P205 Hommel A.  P215 Honecker F.  P269, V107, V878 Hönes J.  V33, V632 Hönes J.M.  V34, V611, V852 Hopfer O.  P477, P779 Hopfinger G.  V835 Hopfner K.-P.  V133 Hoppe S.  V565 Hoppenau C.  P205 Hopprich A.  V731 Horger M.  P198 Horn H.  V421 Horn P.A.  V124, V849, P186 Hornemann B.  P483, P492, P496 Hörnig S.  P463 Horny H.-P.  V581 Horst H.-A.  V32, V424, V657 Hose D.  P773, V40 Hoskin P.  P264, V882 Hoster E.  V421, V71 Hozaeel W.  P214, P827 Hübel K.  P239, P240, V147 Huber A.  V369 Huber E.  P449 Huber M.  P248 Huber U.S.  P260 Huberle C.  V853, V857 Hubert K.  P173, V725 Hubmann M.  V127 Hückelhoven A.  V125 Hudecek M.  P772, V587 Hug M.J.  V752, P486, P504 Hughes T.P.  V600, V604 Huguet F.  V285

Oncol Res Treat 2015;38(suppl 5):271–288

277


CONTENTS AUTHOR INDEX

Hüllein J.  V420 Huls G.  V161 Humblet Y.  P211 Hummel M.  P215, P495, V28, V421, V660 Humpe A.  V36, V165, V561 Hundemer M.  P769, V546 Hunfeld K.-P.  P214 Hurtz H.-J.  P256 Hust M.  P781 Huth A.  V618 Hutson T.  V673 Hutter B.  V420, V687 Hutter G.  P216, P221, P223 Hutterer E.  V664 Hüttmann A.  V869 Hutzschenreuter F.  V609 Huylebroeck D.  P482, P758 Huynh M.Q.  P474

I Iannazzo S.  P450 Ihle M.  V887 Ihme S.  V559 Ihne S.M.  P237 Ihorst G.  P179, P774, P775, P776, P785, V42 Ikenberg R.  P777, V732 Ilaender A.-K.  V742, V746 Ilariucci F.  V584 Ilhan O.  V291 Illerhaus G.  P222, P224, P780, V302, V584 Illert A.L.  V617 Illig D.  P802 Illmer T.  P255, V115 Imbach P.  V6 Indenbirken D.  V390 Indrak K.  V603 Irrgang P.  P221 Isaakidis K.  V286 Isbell L.K.  P224 Isfort S.T.  P754, P760, P761 Isnard F.  V285 Issels R.  P806 Istvanffy R.  V343, V543, V544, V617 Italiano A.  V579 Izbicki J.R.  P200, P206, P207, P208, P209, P210

J Jabbour E.  V424 Jackisch C.  P823 Jacob U.  V133 Jacobi B.  V130 Jaeger T.  P500 Jaehde U.  P258, V46, V408, V685 Jaenecke C.  V423 Jaeschke B.  P189 Jagannath S.  V548 Jäger D.  V431, V687 Jäger E.  P792 Jäger P.  P174 Jäger U.  V422 Jahn N.  V538 Jahnke K.  P779

278

Jakab A.  V41 Jäkel N.  P173, P241, V725 Jakob C.  P779 Jakob F.  V549 Jakobs D.  V42 Jakubowiak A.J.  V548 James D.F.  V291 Janda A.  V607 Jänicke M.  V45, V75, V394, V395, V670, V674, V886 Janka G.  V851 Jann J.C.  P231, P234 Janning M.  P773 Jansen C.  P258 Janssen J.  P256, P456, P458 Janzen V.  P180, P482, P758 Jaquiéry C.  V615 Jaroslaw M.  V611 Jarzab B.  V93, V613 Jaschonek K.G.  P768, P788 Jassem J.  V673 Jauch A.  V40 Jawhar M.  V581, V724 Jedraßczyk M.  P507 Jedrzejczak W.W.  V73 Jeffers M.  P201 Jehn C.  P779, V859 Jehn L.B.  P178 Jelkmann W.  P817 Jensen B.  V657 Jensen I.S.  P517 Jentsch-Ullrich K.  P473, V81, V605 Jentzsch M.  P187, P241, V43, V126, V428, V541 Jeremias I.  V134, V715 Ji S.  V28 Jilg S.  V853 Jitschin R.  P181, V691 Jochum C.  V72 Joensuu H.  P803 Joerger M.  V46, V612, V614 Johnson P.  V73, V584 Jöhrens K.  P495 Jokic M.  V557 Jonas D.  P504 Jones J.A.  V291 Jongen-Lavrencic M.  V307 Jordan K.  V629 Jorg T.  P493 Jørgensen J.  P227 Jost E.  P761, V345 Jost P.J.  V299, V853 Josting A.  P779 Jotterand M.  V601 Jülicher H.  V533 Jumaa H.  V367 Jung B.  P189 Jung G.  V846, V850 Jung M.  P171, P504, V42 Jung R.  P782 Jung W.  V285 Jungberg P.  P255 Junghanß C.  P455, P505, P796, V81, V285, V288, V301, V423, V734, V735, V747 Jurczak W.  V73 Jurk K.  P819

Oncol Res Treat 2015;38(suppl 5):272–288

K Kaemmerer D.  Kaestner L.  V351 Kahl B.S.  V73 Kahn S.  P472 Kähnert H.  V568 Kahrs A.-K.  V561 Kaifie A.  P754, P761 Kaina B.  V306 Kaiser F.  P507, P514, P801, P802 Kaiser G.  V25, V27 Kaiser S.  P486, P504, V752 Kaiser U.  V418, V419, V426 Kalanovic D.  P226, P266, P267 Kalb B.  V557 Kale J.  V309 Kalliara I.  P825 Kalmanti L.  V601 Kaminsky B.  V887 Kämmerer D.  P461, P476, P495, P828 Kampa-Schittenhelm K.  P172, P176, P184, P185, P212, V540 Kämpfe D.  V80 Kampmann E.  P806 Kang Y.-K.  P803 Kantarjian H.M.  V84, V85, V424, V604 Kanz L.  P172, P176, P184, P185, P198, P212, P444, P472, P763, P766, P768, P788, V346, V430, V434, V539, V540, V550, V564, V602, V661, V662, V686, V688, V744, V846, V850, V858, V861 Kanzler S.  V392 Kappeler C.  P803, V93, V613 Kapp-Schwörer S.  V32 Karatas A.  P468 Karsten A.  P237 Karthaus M.  P201, V618 Kase J.  V660 Kasenda B.  P780 Kasheverov I.  V878 Kasper B.  V105 Kasper S.  V25, V27 Kast K.  V730 Kauff F.  V418, V419 Kaufmann M.  V426 Kaumanns A.  V32 Keating M.  V291 Keil F.  P227, P229 Keilholz U.  V391 Keller A.  P755, V42 Keller R.  V392 Keller U.  V308, V715, V718 Kellner C.  V36, V561 Kemele M.  V302, V304, V704 Kempf V.A.J.  V704 Keppler U.  P480 Kerkmann M.  V629 Kern W.  V720 Kerstan A.  P237 Kerstan H.  P762 Keye P.  V537 Khan S.A.  V563 Khandanpour C.  V33, V34, V611, V632, V852 Khoder N.  V43 Khoury H.J.  V604

Author Index


CONTENTS AUTHOR INDEX

Kiani A.  V426 Kiecke C.  V305 Kiefer-Trendelenburg T.  P455 Kiehl M.G.  V426, V625, V618, P477 Kiesewetter B.  P476, V422 Kiewe P.  P779, V81 Kiladjian J.J.  V721 Killer H.  P238 Killer M.C.  P765 Kim D.-W.  V604 Kim H.-J.  V673 Kim S.-B.  V91 Kim Y.  V293 Kimmich M.  V46 Kindler T.  V32, V35, V306 Kingreen D.  P779 Kirchhoff A.  V672 Kirchner E.  V620 Kirchner H.  V425, V675 Kirschbaum B.  P796 Kirschbaum R.  V722 Kischel R.  P175, V134 Kisro J.K.  V81 Kiyota N.  V91 Klade C.  P759 Klapper W.  P220, P478, V299, V302, V421 Klare P.  P255, V432 Kläsener K.  V537 Klatt S.  V341, V719 Klausmann M.  P494, P754, V350 Klausz K.  V561 Klawitter S.  P473 Kleber M.  V378 Kleboth K.  V433 Klein A.  P799, V750 Klein B.  P773 Klein M.  V426, V625 Klein S.A.  V862 Kleiner H.  P454 Klenner A.  P436 Kleylein-Sohn J.  P494 Klier J.  V533 Klimek V.M.  V424 Klingeberg C.  V617 Klingner K.  P224 Klink A.  P244, P442 Klobuch S.  V562, V692 Kloss S.  P262 Klotz M.  P192 Klug J.  P486 Kluge A.  P466 Kluth S.  V290, V292, V580 Kluth-Pepper B.  P823 Knauf W.  P783, V75 Kneba M.  V71, V287, V289, V292, V602 Knecht H.  V287 Knecht R.  P822 Kneifel S.  P821 Kneis S.  V742, V746 Kneitz C.  P791 Knittel G.  V663 Knop S.  P237, P772, P778, P787, V39 Knorn A.-M.  V299 Knösel T.  P806 Knyrim M.  V126, V428, V541

Author Index

Ko Y.-D.  V38, V46, V432, V685, V885 Kobbe G.  P174, P233, P247, P251, P271, V116, V161, V753 Köberle D.  P467 Kobina S.  P201 Kobold S.  V24, V133 Koch A.  V672 Koch K.  V421 Koch L.  P258 Kochkorov A.  P238 Köchling G.  V81 Koehler M.  P508, V565, V630 Koerner S.  Kofla G.  V618 Köhl U.  V709 Kohlbacher O.  V550, V686 Kohlbrenner K.  V602 Köhler A.  V294 Köhler P.  P797, P798 Kohlweyer U.  V42 Köhne C.-H.  V32, V601 Köhnke T.  P170, P175, V127, V131, V134 Kolanus W.  V723 Kolbe-Busch S.  P247 Koldehoff M.  P242, P438, V162, V625 Koller E.  V425 Komarica V.  V716 Konantz M.  V342, V434, V539, V582 Kondakci M.  P233, P247, P250, P251, V753 König F.  V554 König K.  V885 König V.  V88, V402 Königsmann M.  P252, V81 Königsrainer A.  P252, V686 Konkolefski C.  V288 Koopmann J.  V287 Kopfmann S.  V26, V394, V395 Kopp H.-G.  P198, P763, P766, P768, V46, V662, V688 Köppler H.  P820, V433, V748 Koptina A.  P818 Korf U.  P205 Korfee S.  P491 Korfel A.  P215 Korger M.  P453 Körmöczi U.  P437 Kornberger T.  P229 Körner S.  V846, V850 Körper S.  V128 Korsching E.  P805, P807 Kortüm K.M.  V551 Koschmieder A.  V426 Koschmieder S.  P754, P756, P757, P760, P761, V426 Koschny R.  V74 Koska M.  V674 Kostenko A.  V884 Köster R.  V33, V34, V632 Koukakis R.  V684 Kousis P.  V661, V850 Kovacs G.  V290, V666 Kowalewski D.J.  P472, V550, V564, V686 Kraff S.  V46, V685 Kragl B.  V81 Krahl R.  P173, V43, V725

Kralj M.  P437 Kramer M.W.  V880 Kramer M.  V730, V855 Krämer A.  P497 Krämer I.  V74 Krammer-Steiner B.  P194, P196, P791 Kraus S.  V549 Krause M.  V583 Krause R.  P245 Krause S.W.  V426, V601, V602, V625, V691 Krause T.  V634 Krauß U.  V346 Krauter J.  V425, V538, V714 Krebs L.  V135 Kreher S.  P215 Kreil S.  V862 Kreisselmeier K.-P.  P766, P768 Krekeler G.  P226, P266, P267 Kremer A.  P816 Krenn P.  V664 Kreppel D.  V132 Kreutmair S.  V617 Kreutz M.  P816 Kreuzer K.-A.  V290, V292, V609, V665, V666 Kriege O.  V35 Krieger S.  V32 Kriegs M.  V390 Kriegsmann M.  P497 Kriesen U.  V734, V735 Krippl P.  P453 Kritikou P.  V732 Kröger N.  P773, P781, V161, V627 Kron F.  V884 Kröning H.  P213, V682 Krönke J.  V32, V39 Kroog G.  V83 Kropf S.  V630 Kropp K.N.  V688 Kropshofer H.  P440 Kroschinsky F.  V641 Krtschil M.  P491 Krug U.  V424, V426 Kruger S.  V127 Krüger M.  V665 Krüger S.  V887 Krüger W.  P436, P445, V429 Kruggel L.  V670 Krumbholz A.  V165 Krumm K.  P473 Krupka C.  P175, P493, V134 Kuball J.  V125, V161 Kube D.  P478, V706 Kuchenbauer F.  V39 Kuczyk M.A.  V880 Kudryavtsev D.  V878 Kuebler A.  V850 Kuepper M.K.  V307 Kufer P.  P175, V134 Küffer S.  V305 Kuhn A.  V616 Kuhn M.  V37 Kuhn W.  P258 Kühne R.  V881 Kull M.  V39

Oncol Res Treat 2015;38(suppl 5):271–288

279


CONTENTS AUTHOR INDEX

Kunde-Krüger J.  P487 Kündgen A.  P233, V32, V425, V538 Kundt G.  P796 Kunecki J.  V620 Kunitz A.  P770 Kunz C.  V40, V615 Kunz K.  V35 Kunzmann V.  V38, V426, V685 Küppers R.  V365, V704 Kurbacher C.M.  P255, P256 Kurts C.  V848 Kuru T.H.  V883 Kurutz J.  V299 Kuss I.  P803 Kutscheidt A.  V682 Kuzmina Z.  P437 Kvalheim G.  V131 Kvasnicka H.-M.  V80 Kwiatkowski M.  V369

L La Rosée P.  P447, V584, V851 Labar B.  V118 Lammert H.  P495 Lampe H.  P796 Lams R.F.  V33, V632 Lancet J.E.  V424 Landwehrmeyer B.  V620 Lang A.  P453, P500 Lang F.  P754 Lang P.  P493, V708 Lange C.  P262 Lange E.  P456, V605, V714 Lange T.  P458, V126, V428, V541 Langer C.  V39, V547 Langer F.  V413 Langerak A.W.  V564 Langerbeins P.  V290, V292, V666 Langguth P.  P794 Lantuejoul S.  V888 Larkin J.  V673 Laryionava K.  P792 Lass Flörl C.  P798 Lassmann S.  V616 Lathan B.  P456, V606 Laurent D.  P201 Lauseker M.  V601, V602 Lauten M.  V626 Le Beau M.M.  V119 Le Cesne A.  V579 Le Coutre P.  P450, P452, V83, V604, V605, V859 LeBeau M.  V114 Leber B.  V309 Lechner A.  V94 Lechtape B.  P807 Ledderose G.  V127 Lee S.  V28 Lehmann N.  V306 Lehmberg K.  V851 Lehners N.  V74 Lehrnbecher T.  V626 Leibbrand B.  V568 Leiblein S.  P241, V43, V126

280

Leibold J.  V850 Leich E.  V421 Leichtle R.  P480, V350 Leithäuser M.  V734, V735 Leitner C.  P500 Lellek H.  V627 Lemmermann N.  V562 Lenard A.  V582, V633 Lengerke C.  V161, V342, V430, V434, V539, V582, V633, V858 Lengfelder E.  P183, V526 Lennartz K.  V34 Lenz F.  P483, V730 Lenz H.-J.  P201, P211 Lenze D.  P215, V421, V660 Lepetre S.  V285 Leppä S.  P227 Lerchenmüller C.  P473 Leser U.  V660 Lestin M.  P194, P196, P791 Leuteritz K.  V400 Levis A.  V114 Li J.  P482, P758 Li Y.  V291 Li Z.  V308 Lichtenegger F.S.  P175, V131, V134 Liebregts T.  V625 Liebs S.  V391 Liedgens P.  V663 Lienhard R.  P182 Lilly M.A.  V426 Lin C.-C.  V132 Lindberg E.  V852 Linde H.  P451, P779, V295, V684 Lindemann M.  P438, V124 Lindemann H.-W.  V40, V605, V714 Lindig U.  V392 Lindner D.  V125 Lindner L.  P493, P806 Linge A.  V583 Link H.  P252, V426, V629, V650 Link K.  V685 Linke F.  P478, V706 Linke R.  V672 Linton K.M.  V584 Lion T.  P448 Lipp P.  V351 Lipp R.  P510, P515, P518 Lippl S.  V127 Lisec J.  V660 Lisenko K.  P228, P243, P769, V546, V659, V863 Liss B.  P798 Lissandre S.  V285 Loewecke F.  V299 Löffler C.  P795 Löffler H.  P497 Löffler J.  P795 Löffler M.W.  V686 Loges S.  P773, V390 Lohaus F.  V583 Lohneis P.  P826, V28, V303 Lohoff M.  P474 Loibl S.  V432 Lollert A.  V115

Oncol Res Treat 2015;38(suppl 5):272–288

Lonial S.  V548 Lopez Niedenhoff D.  P251 Lorch A.  P271 Lordick F.  V22, V360, V696, V733 Lorenz A.  P255 Losem C.  P213, P235, P256, P452, V418, V419 Lotfi S.  V724 Lotze C.  P491 Lubberich R.K.  P756 Lübbert M.  P171, P179, V114, V118, V119, V161, V425, V538, V714 Lück A.  P252, P256, P473, P519, V115, V682 Lück H.-J.  V432 Lüdecke G.  V554, V675 Lüder F.  P779 Ludescher C.  P453, P767 Ludwig H.  V97 Lüers A.C.  P460, P462, P464, P466, P468, V47 Luft T.  P439 Lüftner D.  P823 Lupp A.  P461, P476, P828, V435, V671 Lustgarten S.  V600, V604 Lustig D.  P446

M Ma T.  V714 Maas C.  P252 Maas-Bauer K.  V161 Maccari B.  V125 Mach N.  V90, V92 Machein M.  V752 Machtens S.  V52 Maciejewski J.  V852 Macintyre E.  V71 Mack S.  V620 Mackensen A.  P181, P230, P816, V300, V552, V691, V860 Mackensen F.  V847 MacKenzie C.  P247, P251 Madanchi R.  P269, V878 Mades A.  V563 Maecker-Kolhoff B.  V709 Maerz W.  V555 Magalhaes S.  V114 Mahon F.-X.  V83 Mai E.K.  V38, V40 Maier B.  P769 Maier V.  P502 Maintz C.  P754, P783 Makarieva T.  V878 Maki R.  V579 Makishima H.  V611, V852 Malchau G.  P784 Malcovati L.  V114 Mallela N.  P805 Manka L.  V369 Manos G.  V85 Manz P.  V346, V545 Märker-Hermann E.  P189 Markert U.R.  V435, V671 Markhauser M.  P757 Märklin M.  P763, V662

Author Index


CONTENTS AUTHOR INDEX

Markmann S.  P796 Marks R.  P219, V161, V658 Markus M.  V25, V27 Markus P.  V25, V27 Marneth A.  V852 Maroto P.  V673 Marretta L.  P790 Marschner N.  V26, V45, V75, V394, V395, V670, V674, V886 Martens H.  V80 Martens U.M.  P511, V392 Martin P.  V73 Martin R.  V854 Martin V.  V90, V92 Martinez C.  V164 Marx A.H.  P200, P206, P207, P208, P209, P210 März M.  V390 Maschmeyer G.  P779, V418, V851 Massberg S.  V343 Maßwig S.  V303 Mato A.R.  V291 Matschke J.  V72 Matutes E.  V420 Matzdorff A.  V412, V721 Mau-Holzmann U.  V540 Maurer C.  V290, V292 Maurer H.  P179 Maurer J.  V615 Maury S.  V285 Maus R.  V533 May A.M.  P774, P785, V42 Mayer F.  V46 Mayer J.  V83, V85, V683, V429 Mayer K.M.  P180, P246, P254, P257 Mayerhoefer M.E.  V422 McClanahan F.  V659 McGinnis C.  V633 Meckel K.  P503 Medinger M.  V41, V858 Meggendorfer M.  V720 Mehnert A.  V733 Mehnert K.  P491 Mehta C.  V424 Meier F.M.  V672 Meiler J.  V25, V27 Meincke M.  P452, P456 Meisel C.  V634 Meiß F.  V752 Meissner T.  P773 Meissner-Weigl J.  V549 Meister R.  V748 Mejstrikova E.  V607 Melichar B.  V673 Mellado B.  P264, V673, V882 Melling N.  P200, P206, P207, P208, P209, P210 Mendoza-Schulz L.  V751 Menon R.  V888 Menschel E.  P229 Mensen A.  V634 Merkelbach-Bruse S.  V49, V884, V885, V887 Merker N.  V625, V626 Mernberger M.  P203 Mertens T.  V125

Author Index

Mertins S.  P808 Merz B.  V430 Merz M.  V40 Merz W.M.  P180 Mesa R.A.  P757 Metzelder S.K.  V161, P178 Metzgeroth G.  V581, V724 Metzler G.  P198 Mewes H.W.  V544 Meyer B.  P455 Meyer E.  V161 Michaelson M.D.  V673 Michalski J.M.  P261 Michel C.  V743 Michel L.C.  V33, V34, V611, V632, V852 Michels B.  V125 Michels S.  V49, V884, V885, V887 Middeke M.  V163 Mies A.  V429 Miethe S.  P781 Mikesch J.-H.  V717 Milani V.  V567 Milazzo A.  P212 Miller K.  V879 Miller M.C.  V46, V685 Millis S.Z.  V556 Milpied N.  V164 Min D.  V344 Minck C.  P831 Mineur L.  P211 Mingrone W.  V881 Mir P.  V430 Mischler K.  P821 Mitchell S.A.  P489 Mitterer M.  P453 Mlineritsch B.  V31 Möbus V.  V432 Moch H.  V434 Mochmann L.H.  V286 Mohamed H.  V84 Mohapp A.  P490 Möhle R.  P444, V346, V722, V861 Mohm J.  V75 Mohr A.  V721 Mohr B.  V855 Mohring M.  P223 Mohty M.  P240, V164 Molina A.  V673 Molinari F.  V90, V92 Molitor E.  P246 Moll R.  P203 Möller A.-K.  P441 Möller M.-D.  V42, P774, P785 Möller P.  V421 Molnar I.  V93, V613 Mönnich D.  V583 Monoranu C.-M.  V302 Monsef I.  P239, V609 Monteverde S.  V121 Montoto S.  V164 Moosmann A.  V131 Morant R.  P260 Moreau P.  V548 Moritz B.  V46, V685 Morlot S.  V326

Moroy T.  V611, V852 Mossner M.  P231, P234, V286 Möstl M.  P229 Motzer R.  V673 Mougiakakos D.  P181, V300, V691 Mrachacz H.  V43 Mück F.  P444 Mügge L.-O.  P442, V39 Muggen A.F.  V564 Mühlenberg T.  P438 Müller A.M.  V344 Müller F.  P797, P798 Müller G.  V341, V719 Müller H.  V137 Müller J.  V582 Müller K.  P766 Müller L.  P270, V46, V605, V674, V685 Müller L.P.  P177, P188 Müller M.  V657, V851 Müller M.C.  P452, P454, V84, V600, V601, V602, V604, V626 Müller M.R.  P763, P766, V662 Müller M.J.  P179 Müller T.  P204, P272 Müller-Thomas C.  V119, V718, V853, V857 Müller-Tidow C.  P177, P188, P202, P204, P272, V426, V714 Műzes G.  P475 Mumm F.  P489, P792, V127 Munder M.  V40 Münz M.  V45, V670, V674, V886 Murawski N.  V656 Murga Penas E.M.  P236 Murua Escobar H.  V288, V301, V423 Musch R.  P519, V394, V395 Mußbach F.  P828 Muus P.  V118 Mytilineos J.  P469, V128

N Na I.-K.  V161, V634, V859 Nabian B.  P461 Nagel I.  P236, V289 Nagele E.  P490 Nagorny N.  P251 Narayanan G.  V85 Nauck F.  V733 Naumann N.  V581, V724 Naumann R.  V38, V426 Nazari S.  V533 Neben K.  V38 Neemann N.  P462, P464, V47 Negrin R.  V161 Neidig C.  P235 Nelde A.  V564 Nellessen C.  P180, P254 Neubauer A.  P178, P203, P273, P474, P765, P830, V38, V161, V426, V602, V714 Neuberger C.  P182 Neuhof A.  P226 Neukirchen J.  V116 Neumann F.  V300 Neumann M.  P231, V286 Neumann T.  P436, P445

Oncol Res Treat 2015;38(suppl 5):271–288

281


CONTENTS AUTHOR INDEX

Neumeister P.  P218, P443 Neureiter D.  V664 Newbold K.  V91 Nguyen B.-P.  V553 Nicolini F.E.  V604 Niederwieser D.  P173, P187, P241, V43, V126, V428, V541, V725 Niedtner R.  P266 Niegisch G.  P271 Nietsch J.  V629 Nietzke M.  V732 Nilius V.  P273 Nilsson S.  P261, P264, V879, V880, V882 Nilsson S.K.  V341 Nist A.  P203 Noessner E.  P806 Nogai A.  P770 Nold P.  P765 Nolte F.  P183, P231, P234, V608, V625, V862 Nolting A.-C.  V116 Nolting J.  P829, V552 Noppeney R.  P240, V426 North T.E.  V582 Nösslinger T.  P227, P229 Noureddine R.  V25, V27 Nowak D.  P231, P234 Nowak V.  P231, P234 Nowek K.  V307 Nuber J.  P190, P197 Nusch A.  V670, V674 Nußbaumer E.  V307 Nutting C.  V613

O O’Sullivan J.  V879, P264 Ober A.  V432 Oberg H.-H.  V289 Oberle A.  P781 Obländer J.  P231, P234 O'Brien S.  V291 Obstfelder E.  V619 Oduncu F.S.  V133 Oechsle K.  P822, V110 Oelschlaegel U.  V855 Oette M.  V657 Oexle H.  P453 Oh J.  V561 Öhler L.  P767 Ohyashiki K.  V114, V119 Oing C.  V556 Oldenkott B.  P779 Ollech-Chwoyka J.  P455 Oostendorp R.A.J.  V343, V543, V544, V617, V715, V857, V853 Opalka B.  P438, V34, V632 Opeker K.  V752 Opferman J.T.  V431 Ordemann R.  V161 Orsini L.  V48 Orth M.  V620 Ortiz Tanchez J.  V286 Ortner P.  V629 Osburg S.  V749 Oskay-Özcelik G.  P255

282

Osterborg A.  V291 Ostermann G.  V81 Ostermann H.  P240, P446, P463, P517, V745 Otremba B.  P253 Ott G.  V421, V708 Otte K.  P269, V878 Otte P.  P486, V752 Ottinger H.  V124 Ottmann O.G.  V285, V289, P452 Overbeck T.  P468 Overkamp F.  V675

P Pachmann K.  P502, V631 Pachmann U.  P502, V631 Pacini F.  V613, V93 Paczulla A.  V539 Pagel C.  V343, V543, V544, V715, V857 Pagel J.  V293 Pahl A.  V746 Pahl H.L.  P757 Pall G.  P500 Pandurevic M.  P774, P785 Panny M.  P227, P229 Panse J.  P232, P782, V285, V350, V618, V851 Pantel K.  P773 Pantic M.  P774, P775, P776, V42 Papachristofilou A.  V106 Papasotiriou I.  P825 Papazoglou D.  V881 Pape V.  V305 Papke J.  P255 Paquette R.  V604 Parampalli Yajnanaraya S.  P813 Pareigis S.  P487 Parker C.  P261, P264, P265, V880, V882 Parmentier S.  P754, V635 Parren P.W.H.I.  P793, V561 Paschka P.  V32, V425, V538 Paschke R.  V93, V613 Pasemann S.  V300, V552 Pass D.  P493 Passweg J.  V41, V161, V858 Pastuschek J.  V671 Patel S.  V579 Patella F.  V310 Paul A.  V25, V27 Paul S.  P188 Paul U.  P236 Paula H.  V410 Pauligk C.  P214, P827 Pavel M.  P498 Pavel P.  P243, V546, V863 Peceny R.  V426 Pecher A.-C.  P768, P788 Pecherstorfer M.  P767 Peipp M.  V36, V561 Pelzer U.  P826 Penrod J.R.  V48 Penter L.  V161 Peplinski D.  V565 Pereboom T.  V430 Perner S.  V430, V434

Oncol Res Treat 2015;38(suppl 5):272–288

Peschel C.  V132, V161, V299, V308, V343, V543, V544, V547, V715, V718, V853, V857 Peter B.  P755, V621 Peter N.  V38 Peters E.  P516, V751 Petersen V.  V886 Peterson L.  P446 Peters-Regehr T.  V40 Petrides P.E.  P759, V359 Petsch S.  V567 Petzer A.  P453 Pews-Davtyan A.  V301, V423 Peyn A.  V38 Peyrade F.  V420 Pezzutto A.  P770, V542 Pfarr N.  P497 Pfarrer C.  V671 Pfeffer K.  P250, P251 Pfeifer D.  V616 Pfeifer H.  V285, V289 Pfeifer M.  P191, P192 Pfeilstöcker M.  V114, V119, V274 Pfirrmann M.  V601, V602, V603, V605 Pflug N.  V292, V580, V666 Pflüger K.-H.  V118 Pfrepper C.  V43 Pfreundschuh M.  P191, P192, V299, V302, V304, V362, V601, V602, V656, V704 Pfründer D.  P253 Philip B.  V293 Philipp-Abbrederis K.  V718 Photini S.M.  V671 Pichler M.  P218, V307 Pickl W.  P437 Pieger R.  V620 Pietschmann E.  V308 Pietzonka S.  P254 Pigneux A.  V424 Pigorsch S.  V583 Pinilla-Ibarz J.  V604 Pink D.  P436 Pircher A.  V420 Piribauer M.  P789 Pizon M.  P502, V631 Plachter B.  V562 Plath M.  P253 Plattfaut C.  V689, V690 Platzbecker U.  P754, V37, V115, V118, V119, V163, V276, V350, V429, V852, V855 Platzek I.  P810 Pleß M.  V126 Plötze M.  V43 Pobiruchin M.  P511 Podar K.  V431 Podlech J.  V562 Podleska L.-E.  P804, P808, V553 Poeck H.  V132 Poeschel V.  V656 Poettler M.  P499 Pohlkamp C.  P779 Politi L.S.  V584 Poljak A.  P763 Poller K.  P816 Pomplun C.  P236

Author Index


CONTENTS AUTHOR INDEX

Pönisch W.  P187, P241, V43, V126, V428 Ponzoni M.  V584 Popescu R.A.  P260 Porowski P.  P473 Port M.  V350 Postina P.  V626 Potenberg J.  P831, V672 Potratz J.  P805, P807 Pott C.  V71, V287 Pöttgen C.  P804, P808 Potthoff K.  V847 Potzner M.  P465, V889 Prager G.W.  P499 Prange-Krex G.  P473, V115 Prazeres da Costa O.  V544 Prejzner W.  V603 Prenzel R.  P462, P464, P466, V47 Pressler J.  P231, P234 Preukschaß M.  P217 Preuss K.-D.  V302, V304, V704 Prevalsek D.  V127 Prochazkova J.  V429 Proetel U.  V602 Prokein A.  V620 Promny I.  P775 Prüfer S.  P819, V743 Pucher S.  V664 Pukrop T.  P205, V584, V706 Pulczynski E.  V584 Pulewka K.  V566 Puppe B.  V421 Pursche B.  P218 Puthenparambil J.  V546

Q Quast T.  V723 Quecke T.  V689, V690 Quehenberger F.  P245 Quidde J.  P214 Quintanilla-Martinez L.  V539

R Raab M.S.  P773, P777, V548, V585 Rachlis E.  P251, V753 Rachow T.  P503 Račil Z.  V429 Raderer M.  P476, P498, V422, V591 Radford J.  V73 Radiovoyevich T.  V852 Radke J.  P810 Radke S.  P463, P468 Radsak M.P.  V743, P794, P819 Raffoux E.  V285 Rahn S.  P250 Raić A.  V345 Ramachandran A.  P176 Rammensee H.-G.  P472, V550, V564, V686 Randazzo M.  V369 Rao A.  V662 Rao V.  V537 Rapp M.  V24 Rasche L.  P778 Rassaf T.  V753

Author Index

Rath P.M.  V625 Rauch D.  P260 Rauh J.  P252, V75 Rautenberg B.  V752 Ravandi F.  V424 Rea D.  V84, V604 Rebmann U.  P266, P267 Rebmann V.  V124 Recher C.  V424 Rechkemmer S.  P170 Reck M.  V46, V48, V888 Recker F.  V369 Reddehase M.  V562 Redlich K.  P499 Reece D.E.  V548 Regitz E.  V302, V304, V704 Rehe K.  V692 Reich S.  V434 Reichardt P.  P803 Reichart A.  P214, P827 Reichert D.  P452, V684 Reichle A.  V285, V426, V692 Reid G.  V35 Reidel V.  V853 Reim R.  V722 Reimann C.  P461 Reimann M.  V303 Reimer T.  P796 Reinacher P.C.  P224 Reinbach M.-C.  V753 Reinecke P.  P232 Reinhardt H.C.  V557, V558, V663 Reinhardt H.  P486, P504, P785, V42, V752 Reinhardt M.  P466 Reinhardt P.  V128 Reinholz U.  V731 Reinmuth N.  P468, V46 Reinwald M.  V625, V626, V862 Reipsch F.  P204 Reis A.-C.  P804, P808, V553 Reischl J.  P201 Reiser M.  P253, P451, P473 Reitan J.  P240 Reiter A.  V581, V722, V724 Reni M.  V584 Rentsch A.  P483 Rentsch C.  P260 Repp R.  P451, V426 Reth M.  V537 Reuning-Scherer J.  P265 Reuss-Borst M.  P516, V751 Reuter S.  P819 Rexa B.  P455 Rexrodt P.  P195 Reyher-Klein S.  P831 Rha S.Y.  V579 Rhein C.  V292 Ribera J.-M.  V285 Ribrag V.  V71 Richardson P.G.  V548 Richner J.  V881 Richter C.  V547 Richter D.  V565, V687 Richter S.  P810 Rick O.  V87

Riecken K.  P773, V390 Riedel J.  V734 Riedner C.  V567 Riedt T.  P482, P758 Rieger C.  P446, V745 Rieke J.  P459 Riera Knorrenschild J.  V392 Riese C.  P799 Riess H.  P826 Riess O.  V555 Riester A.  V620 Rieth A.  V684 Riethausen K.  V82 Rietkötter E.  V610, V854, V856 Rinaldetti S.  V602 Rinas N.  V567 Rinderer F.  P500 Ringhoffer M.  V32, V429 Rinke J.  V619 Ritchie E.K.  V424 Rittig S.M.  P812, V688 Rivera V.M.  V600, V604 Rizzi M.  V607 Robinson B.  V91 Robinson S.  V164 Roboz G.J.  V424 Rochau U.  P453 Rödel C.  V583 Roder H.  P242 Roder J.  P242 Rody A.  V690 Roeder I.  V37, V385 Roessler M.  V46, V685 Rogers S.  V324 Roggenhofer S.  P509 Rohde C.  V426 Rohde H.  V627 Rojewski M.  V559 Rolfs A.  V301, V423 Röllig C.  V37, V426, V429, V637 Romaguera J.E.  V73 Römer S.  V735 Roolf C.  V288, V301, V423 Roos W.P.  V306 Rosenberger A.  P815 Rosenwald A.  V302, V421 Roskopf C.C.  V133 Roskos M.  V725 Rösler W.  V691 Ross R.S.  P438 Rotermund M.  P764 Roth A.  V80 Roth B.  P786 Roth P.  P215, V302 Röth A.  P479, V350, V584 Rothaug W.  P790 Rothermundt C.  V67, V881 Rothfelder K.  V850 Rothfuss O.  V430 Rothmann F.  P779, V851 Rothschild S.I.  P260, V44, V94, V881 Rottal A.  P437 Rötzer I.  P214 Rousselot P.  V84, V285 Rowlings P.  V83

Oncol Res Treat 2015;38(suppl 5):271–288

283


CONTENTS AUTHOR INDEX

Rubanov O.  P519 Rübben H.  P259 Ruch M.  P486, V752 Rücker F.G.  V716 Ruckert C.  V627 Rudelius M.  V299 Rudolph C.  V425 Rudolph J.  V723 Ruetgen B.  P223 Ruf F.  V543, V544 Rüffer J.U.  V567 Ruhstaller T.  V404 Rule S.  V73 Rumkamp T.  V305 Rummel M.  V294, V418, V419 Rummelt C.  V537 Ruppenthal S.  P454, V351 Ruschpler E.  P241 Russell K.  V556 Rüter B.H.  V118 Rutstein M.  P201

S Saad F.  V879 Saglio G.  V83, V84, V85 Sahakyan L.  P225 Saharyan A.  P225 Sahin U.  P827 Sailer F.  P511 Salamone S.J.  V46, V685 Salat C.  P255 Salih H.R.  P472, V118, V425, V550, V661, V688, V714, V846, V850 Salinas-Riester G.  V854 Salitzky O.  P184 Salles G.A.  V293 Sallmann D.  P779 Salvucci M.  P457 Salwender H.J.  V38, V40 Salzer U.  V607 Salzmann-Manrique E.  V860 Sancho A.  V549 Sandner R.  P256, V45 Sänger J.  P461, P828 Sankot J.  P811 Sanz G.  V114 Sanz M.  V164 Sartor O.  P261, P264, P265, V880, V882 Sasca D.  V35, V306 Sattler M.  V431 Sauer A.  P456, P452, V606 Saur S.J.  P763 Saußele S.  V601, V602, V605 Sauter G.  P200, P206, P208, P209, P210 Savulsky C.  P824 Sawall S.  P773 Sayar H.  V424 Sayer H.G.  P244, P442 Schaab R.  V610 Schaefer C.  P805, P807 Schaefer H.  P224 Schaefer H.-E.  P224, V118 Schäfer T.  V430, V434 Schäfer E.  P757

284

Schäfer H.S.  V302, P222 Schäfer K.-L.  P807 Schäfer K.  P803 Schäfer U.  P800 Schäfer V.  V159, V619 Schäfer-Eckart K.  V426, V618, V285 Schaffrath J.  P272 Schaich M.  V635 Schalk E.  P503, P508, V628, V707 Schanz J.  V114, V117, V119 Schardt C.  P452 Scharrer I.  P819 Schauwecker P.  V125, V128 Scheder A.-M.  P171 Scheel A.H.  V44, V49, V884 Scheele J.  P440 Scheffler M.  V49, V884, V885, V887 Scheibenbogen C.  P193, P495, V634 Scheid C.  P784, V38, V40, V161, V166, V722 Scheideler M.  V307 Schelker R.C.  P449, V341, V719 Schemenau J.  P232 Schenk M.  V605 Schenk T.  P447, V605, V851 Scherrer S.  P488 Schetelig J.  V163 Scheuerlein R.W.  P252, P255, P256 Schewe D.  P773 Schieferdecker A.  P781, V390 Schiemann M.  V343, V544 Schiffer C.  V84 Schild H.  P794, P819, V743 Schildhaus H.-U.  P205 Schildmann J.  P496, V296 Schiller G.J.  V424 Schindler R.  P828 Schinke M.  P775 Schirren J.  P271 Schittenhelm M.M.  P172, P176, P184, P185, P212, V540 Schlee C.  V286 Schlegel P.  P493 Schlegelberger B.  V32, V326, V601, V660, V724 Schleich K.  V303 Schleicher J.  P780, V675 Schleithoff C.  P805, P807 Schlenk R.F.  V32, V425, V538, V620, V716, V722 Schlicht E.  P249 Schlichting A.  V23 Schliemann C.  V717 Schliephake H.  V733 Schlitt H.J.  P195 Schliwa T.  V43 Schlosser P.  V714 Schlosser T.  P222 Schlösser H.A.  V44, V94, V166 Schlumberger M.  V91, V93, V613 Schmalbrock L.  V541 Schmatloch S.  V432 Schmickl M.  V132 Schmid C.  V127 Schmid K.  P481 Schmid K.W.  P259, V25, V27

Oncol Res Treat 2015;38(suppl 5):272–288

Schmid M.  P182, V716, V881 Schmid T.  P467 Schmidt A.  P182, V435, V671 Schmidt B.  V350 Schmidt C.  P455 Schmidt E.  V717 Schmidt K.  P513 Schmidt L.  P461 Schmidt M.  P237, V619, V683 Schmidt S.  P453, P813 Schmidt V.  P244, P442 Schmidt-Wolf I.G.H.  P258, V32, V38, V40 Schmitt A.  V125, V546, V557, V863 Schmitt C.  V28, V279, V303, V660 Schmitt J.  P483, P492, P496, P809 Schmitt K.  P761 Schmitt M.  P171, V125, V294 Schmitt-Gräff A.  V161 Schmitz M.  P822 Schmitz N.  V164, V285, V426 Schmitz S.  P235, V533, V606, V750 Schmohl J.  V858 Schmoll H.-J.  P204, P272 Schnaiter A.  P216 Schnallinger M.  P453 Schneeweiss A.  P823, V431, V432 Schneeweiss M.  P448 Schneider C.  P194 Schneider C.-P.  P465 Schneider E.  V567 Schneider S.  P175 Schneider V.  V129, V559 Schneider-Kappus W.  P452 Schneider-Koriath S.  P194 Schneidewind L.  P436, P445 Schnell R.  V394, V395 Schnetzke U.  V427 Schnittger S.  V581, V602, V720, V724 Schnorfeil F.M.  V131 Schöffski P.  V579 Schöhl M.  P777 Scholber J.  V742 Scholl S.  V427, V714 Scholten F.  P213 Scholz C.W.  P459 Schommers P.  V657 Schönberg K.  V82 Schöning T.  P228, V659 Schönland S.  P439, P769, V169 Schönlieb C.  V680 Schönsteiner S.  V620 Schorb E.  P222, P224, V302, V584 Schöttker B.  P827 Schouten H.  V164 Schrader M.  V675 Schrading S.  V883 Schramm W.  P511 Schreck C.  V343, V543, V544 Schreder M.  P772, P778, P787 Schreieck S.  P453, P767 Schreiner L.  P451 Schrenk K.  V427 Schrezenmeier H.  P469, V125, V128, V350 Schrezenmeier J.F.  V303 Schröder C.  V555

Author Index


CONTENTS AUTHOR INDEX

Schroeder M.  P501, P800 Schroeder T.  P174, P247, P251, V632, V644, V753, V852 Schubert A.  P512 Schubert B.  V730 Schubert J.  P823 Schubert K.  V126, V428, V541 Schuck A.  P250 Schuler E.  P233, V161 Schuler M.  P483, P492, P496, P804, P808, V25, V27, V553, V730, V889 Schuler U.S.  V649, V730 Schüler A.  V35 Schüler F.  P455 Schüler J.  P224, V551 Schulte A.  V390 Schulte W.  V887 Schultze A.  V657 Schulz H.  P270 Schulz K.  P762 Schulz L.  V672 Schulz S.  P476, P828 Schulze I.  P258 Schulze M.  P252, P436, P456 Schulze T.J.  P231 Schulze-Osthoff K.  V430, V434 Schulze-Röbbecke R.  P247 Schulz-Fincke J.  P171 Schumacher B.  V25, V27 Schumacher H.  V747 Schumacher M.  P775, P776, V714 Schumacher M.  P254 Schumacher U.  P269, V878 Schumann C.  V608 Schurich B.  V40 Schuster H.  P472, V550 Schütte U.  P829 Schütz F.  P823 Schuurman J.  P793 Schwaab J.  V581, V724 Schwab K.S.  P246, P257 Schwaiger M.  V718 Schwamborn K.  V718 Schwanbeck R.  P793 Schwartz S.  V16, V286 Schwarz K.  V128 Schwarzbich M.-A.  P228, V659 Schwarzenbacher D.  P218 Schwarzer A.  V115, V605 Schwella N.  P190, P197 Schwenke M.  V689, V690 Schwenke S.  P201 Schwind S.  P187, P241, V43, V126, V428, V541 Schwinger U.  P452, P473 Scriba D.  V47, P462, P464, P466 Sebastian M.  P468 Seckinger A.  P773 Seeber A.  P500 Seggewiß J.  P805 Seidel D.  P797, P798 Seidel H.  P201 Seidel W.  V733 Seifart U.  V86 Seifarth W.  P454

Author Index

Seime T.  V307 Seismann H.  P220 Sekeres M.  V114 Sekora A.  V288, V301, V423 Selder R.M.  P774, P785 Selleslag D.  V118 Sensi A.  P457 Serke M.  P468, V885, V887 Serve H.  P171, P754, V37, V426, V429 Severin K.  V533 Shah N.P.  V600, V604, V83, V84, V85 Shaljyan A.  P225 Shan M.  P265 Sharman J.P.  V293 Shayegi N.  V849 Shcherbakova A.  P818 Shen J.  P465 Sherman S.I.  V91, V613 Shimabukuro-Vornhagen A.  V94, V166, V560 Shirneshan K.  V117, V610, V856 Shizuru J.A.  V344 Shong Y.K.  V93, V613 Shore N.  V882 Siano M.  V90, V92, V154, V612, V614 Sicre de Fontbrune F.  V161 Siddiqi T.  V291 Siebels M.  V675 Siebert F.  P789 Siebert R.  P236, V289, V421 Siebert U.  P453 Siehl J.  V657 Siemer S.  V675 Siena S.  P201, P211, V613 Siffert W.  P259 Silkjaer T.  P227 Sill H.  P245, P443, P815, V307 Simanek R.  P229 Simon G.  P207 Simon R.  P200, P206, P207, P208, P209, P210 Simonitsch-Klupp I.  P476, V422 Sindler P.  P811 Singer K.  P816 Singer S.  P809 Singhal A.K.  V548 Sinn B.V.  P826 Sinn M.  P826 Sipos F.  P475 Skjorestad I.  V880 Sklarz L.-M.  V288, V301, V423 Skoetz N.  P239, V609 Slawik H.R.  P253 Slawska J.  V308 Sliwa T.  P453 Slovak M.L.  V114, V119 Smit J.W.  V93, V613 Smith J.A.  V424 Sobrero A.  P211 Socié G.  V161 Sockel K.  V161, V163, V429 Soffietti R.  V584 Sohm M.  P802 Solé F.  V114, V119 Sommer S.  P814

Sommer U.  V732 Sotlar K.  V581 Sourij H.  P443 Soverini S.  P457 Spadaro S.  V752 Späth-Schwalbe E.  P459 Specht E.  P461, P476 Spenke C.  P495 Sperr W.R.  P448 Spiekermann K.  P175, V127, V134, V285 Spielau-Geidies C.  P795 Spies E.  P220 Spiess B.  V625, V626 Spies-Weißhart B.  V427 Spizzo G.  P500 Spoerl S.  V161 Spriewald B.  P230 Spring L.  V45, V886 Springer G.  V81 Springfeld C.  P827 Sprossmann-Günther G.  V672 Spurgeon S.E.  V73 Spyridonidis A.  V161 Stabla K.  P178, P203, P830 Stadtherr P.  V863 Staehler M.  V674 Stahl M.  V403 Staib P.  V605 Stange T.  V37 Stangl T.A.  P248, P485 Stangl W.M.  P248, P485 Stassen M.  P794, P819 Stauber M.  P443 Stauch M.  P494, V418, V419 Stauder R.  V114, V119 Staudinger M.  V36 Steegmann J.-L.  V603 Stefanzl G.  V621 Steffen B.  V426 Steffens C.-C.  P255 Stegelmann F.  P452, V85, V602, V605, V721, V722 Steigerwald A.  P778 Stein A.  P211, V313, V390 Stein H.  V80, V421 Stein P.  P794, P819 Steinau H.-U.  P804, P808 Steinbrunn T.  P786 Steinemann D.  V660 Steiner R.  P194 Steiner T.  P266, P267 Steinmann J.  P440, V625 Steinmetz T.H.  V350, V533, V606, P458 Stelljes M.  V426 Stempelmann K.  V849 Stenner F.  P260, V881 Stenzinger A.  V687 Stern M.  V41 Stern S.  P452 Sternberg C.  V664 Steudel C.  V115 Steurer M.  V420 Stevanovic S.  P472, V550, V564, V686 Stevens-Brogan M.  V291 Stickel J.S.  P472, P812, V550, V564

Oncol Res Treat 2015;38(suppl 5):271–288

285


CONTENTS AUTHOR INDEX

Stickeler E.  V752 Stiewe T.  P203 Stilgenbauer S.  P470, V73, V290, V291, V292, V293, V666, V832 Stimpfl I.  P248 Stoehr A.  V657 Stöhr A.  P231 Stoiber F.  P263 Stollberg S.  P476 Stolte M.  P474 Stölzel F.  V163, V641 Stonik V.  V878 Stötzer O.J.  V685 Straka C.  V39 Strathmann K.  P756 Straub E.  V746 Strauss A.  P261 Strauß B.  V566 Streichert T.  P784 Streubel A.  V80 Strickland S.A.  V424 Striefler J.K.  P826 Strifler S.  P778, P787 Strik H.  V89 Strobach D.  V745 Ströbel P.  V305 Strobl C.  P816 Strölin P.  P264, P265 Stropiep U.  P462, V47 Strunz A.M.  P267 Strunz B.  V661, V688 Strupp C.  P233 Strüßmann T.  V658 Strüwe S.  V747 Stuart R.K.  V424 Stuhlmann-Laeisz C.  V421 Stühmer T.  P786 Stumme H.  V708 Stümpel J.-P.  P241 Stumpp C.  V683 Stuschke M.  P808, V583 Stute N.  V162 Suarez F.  V285 Subar M.  V83 Subklewe M.  P170, P175, P493, P806, V127, V131, V134 Suciu S.  V118 Suckert N.  V542 Sueptitz J.  V884 Sun W.  P817 Šustkova Z.  V429 Sutradhar S.  V889 Sutter U.  V722 Suttmann I.  V685 Sykoutri D.  P499 Szczepanowski M.  P478, V302 Szczudlo T.  V889 Szedlak G.  P248 Szybinski J.  V35 Szymaniak-Vits M.  P486, V752

T Tabakmakher K.  V878 Tabernero J.  P201, P211

286

Tacke D.  V580 Taeger G.  P804, P808 Tahara M.  V91 Taipaleenmäki H.  P773 Talpaz M.  V600, V604 Tam C.  V291 Tammen A.  P793 Tao S.  V732 Targosz B.-S.  V299 Tauro S.  V114 Tausch E.  V292 Taylor F.  V48 Taylor N.  V657 Tebbe S.  P456 Tebinka-Olbrich A.  P512 Teichler S.  P178, P830 Teichmann B.  P235, P255, P256, P783 Tejpar S.  P201 Teleanu V.  V39, V425 Terracciano L.  P200, P206, P207, P208, P209, P210 Terwey T.  V859 Tesch H.  P255, P451, P456, P458, P757, P823 Teschner D.  V743 Tessen H.-W.  P494, V23, V45, V886, V295, V393, V684 Thaler J.  P453 Thallinger G.  P218 Theis F.  P480 Thelen M.  V94, V560 Theobald M.  P484, V35, V130, V135, V306, V425, V562, V563, V731, V743 Theobald W.  V159 Theocharous P.  P227 Theophil F.  V689 Theurich S.  V166 Theurl I.  V163 Thiede C.  V37, V426, V429, V611, V855 Thiele B.  P781 Thiele T.  P436 Thiem U.  V426 Thieme F.  V165 Thieringer F.M.  V615 Thirman M.J.  V291 Thivakaran A.  V34, V632 Thol F.  V32, V352, V425, V538, V638 Thomalla J.  P820, V433, V748 Thomas M.  V733, V889 Thomas R.  V557, V611 Thomas S.  V562, V692 Thomsen A.R.  V551, P785 Thurner L.  V302, V304, V704 Thuss-Patience P.  V405 Tiemann M.  P217, P270, P460, P462, P466, P468, V47, V888 Tillmanns A.  P807 Timme-Bronsert S.  V616 Timmer H.  V682 Timmesfeld N.  P273 Tinhofer I.  V583 Tischer J.  P514, V127, V164 Tölg M.  P252 Toloudi M.  P825 Tomasek J.  V673 Tomka M.  P789

Oncol Res Treat 2015;38(suppl 5):272–288

Tondar S.  V682 Tonelli M.  P457 Torgovnick A.  V557, V558 Trarbach T.  P235, P783, V25, V27, V394, V395 Trautmann F.  P492, P496, P809 Trautmann H.  V289 Trebicka J.  P760, V723 Treckmann J.W.  V553, P808 Trepel M.  P220, P782 Trittler R.  P504 Troppan K.  P218 Troppmair J.  V307 Trümper L.  P478, V119, V299, V305, V377, V656, V706 Trummer A.  V352 Tsamadou C.  P469 Tsao L.C.  V548 Tschanter P.  V426 Tschuch C.  P224 Tucci M.  V879 Tüchler H.  V114, V119 Turkina A.  V603 Twelves C.  P824 Tzalavras A.  P780

U Übner M.  P230 Uhlmann-Nussbaum C.  V881 Ukena D.  P463 Uleer C.  P823 Ullrich E.  V860 Ulrich-Merzenich G.  P818 Ulshöfer T.  P456, P452 Umbach R.  V581 Unseld M.  P499 Unterhalt M.  V71, V421 Urban C.  P490 Urech C.  P488 Ussat S.  V289 V Vach W.  P774, P785 Vadnais C.  V852 Vag T.  V718 Valdix A.-R.  V393 Valent P.  P448, P453, P755, V114, V119, V621 Valenti A.M.  P457 Valerius T.  P793 Vallet S.  V431 Van Cutsem E.  P201, P211 Van de Loosdrecht A.  V114 Van de Winkel J.G.J.  V561 Van der Reijden B.  V611, V852 Van der Velden W.  V161 Van der Wall K.  V301, V423 Van Essen J.  V883 Van Oorschot B.  V733 Van Roye C.  P820, V433, V748 Vanhoefer U.  V390 Varga Z.  V434 Vassen L.  V852

Author Index


CONTENTS AUTHOR INDEX

Vehling-Kaiser U.  P195, P452, P507, P514, P801, P802 Vehreschild J.J.  P797, V580, V625 Vehreschild M.J.G.T.  P797, P798, V580, V618 Velardi V.  V164 Venkataramani V.  V305 Venz S.  V878 Verbeek M.  V161, V718 Vey N.  V424 Viardot A.  V277, V285, V656 Vick B.  V134, V715 Vij R.  V548 Vilne B.  V544 Virchow I.  P259, P804, P808, V553 Vogel W.  P198, P444, V744, V861 Vogelhuber M.  P449, V692 Vogelzang N.J.  V880, V882, P261 Vogl U.M.  P767 Vogler B.  P782 Vogt J.  V733 Vöhringer M.  V708 Voigt M.  V390 Voigtländer M.  P782 Volegova-Neher N.  V742 Volk H.-D.  P193, V634 Von Amsberg G.  P269, V878 Von Bergwelt-Baildon M.S.  V44, V94, V166, V560, V580 Von Bonin F.  V706 Von Bonin M.  V163 Von Bonin S.  V641 Von Bubnoff N.  P224, V161, V537 Von Burg P.  V881 Von der Schulenburg J.-M.  P513 Von Grünhagen U.  V418 Von Harsdorf S.  P489 Von Kalle C.  V420, V687 Von Lilienfeld-Toal M.  P503 Von Minckwitz G.  V432 Von Müller L.  V704 Von Olnhausen A.  P507 Von Pawel J.  P465, V46 Von Rohr L.  V881 Von Rudloff L.  P801 Von Tresckow J.  V290, V666 Von Verschuer U.  V45 Vonk R.  P201 Vonnahme M.  P760 Vornanen M.  V704 Voskova D.  P453 Voss A.  P500, V556 Vucinic V.  P187, P241, V43, V126, V428 Vuong L.  V859

W Wachsmann G.  V432 Wächte M.  P770 Wachter A.  P205 Wagner A.  P201, P211 Wagner B.  V131 Wagner R.  V426 Wagner S.  V686 Wagner W.  P756, V345 Wahba M.  V882

Author Index

Wahlers K.  P797, P798 Waizenegger J.S.  P773 Walawgo T.  V749 Walch A.  V308 Waldau A.  V619 Walder A.  P453 Waldschmidt J.M.  V42, V551, P774, P775, P785 Walenda T.  P756 Wallau A.  P254 Waller C.F.  P452, P468, V602 Walter A.  P171 Walther R.  V878 Walz I.  V746 Walz S.  P812, V550 Wang H.  V430, V434 Wang J.  V351 Wang L.  V125 Wang M.L.  V73 Wang S.-Y.  P187, P241, V725 Wannesson L.  V881 Ward R.  V424 Wardelmann E.  P202 Warnatz K.  V607 Wartenberg M.  V421 Wäsch R.  P774, P775, P776, P785, V96, V42, V161, V551 Wasmuth J.-C.  V657 Wass M.  P177, P188 Wattad M.  V32, V425 Weber A.N.R.  V564 Weber A.-K.  P794 Weber D.  V32, V425, V538 Weber I.  V135 Weber K.  V23 Weber M.  P484, V731 Weber S.  V731 Weber T.  V851 Wedeken K.  P462, P464 Wedel S.A.  V882 Wehler T.  P484 Wehmeyer J.  P235 Wehr C.  V607 Wehrle A.  V742, V746 Wehrle J.  P757 Weichert W.  P497, V583, V687 Weickert M.-T.  V715, V857 Weide R.  P255, P256, P820, V433, V748 Weidmann E.  V418, V419 Weidner H.  V126, V541 Weigl R.  P437 Weiglein T.  P514 Weiligmann C.  V115 Weinberg K.  V344 Weinhold N.  P773 Weise M.  V641 Weisel K.C.  V40, V550, V588, P768 Weiss H.  P789 Weiss L.  V31 Weissbach L.  V371 Weißenborn G.  P255, P256 Weitz J.  P810 Weller M.  P215, V153, V302 Welslau M.  P799, V294, V418, V419 Weltermann A.  P453

Wendtner C.-M.  V290, V292, V420, V618, V666 Wenge D.V.  V717 Wenk C.  V857 Wennhold K.  V560 Wenzel F.  V545 Wenzl K.  P218 Wermke M.  V163 Werner-Klein M.  V28 Wesselmann J.S.  V666 Weßendorf S.  V714, V392 Wester H.-J.  V718 Westermann J.  V425 Wetzel N.  V395 Wey D.  V748 White D.  V548 Wicki A.  V331 Wickstroem S.  V664 Wider D.  V42, V551 Widmann T.  P509 Widmark A.  P264 Wiedenmann B.  P494 Wiegele K.  P490 Wieschermann U.  P501, P800 Wieser R.  V307 Wiesholzer M.  P453 Wiesneth M.  V125, V128, V129 Wiest G.  P460 Wiesweg M.  V25, V27 Wijermans P.  V118 Wild J.  V661 Wildenberger K.  V126 Wilhelm M.  V685 Wilk M.  V545 Will S.  V625, V626 Willborn K.C.  P462, P464, V47, P466 Willenbacher E.  P500 Willenbacher W.  P500 Williams B.  V341 Williams M.E.  V73 Wilting J.  P478, V706 Wimberger P.  P823, V732 Windemuth-Kieselbach C.  P213, P262 Winkelmann N.  V619 Winkler E.  P792, V687 Winterhalder R.  V881 Wintges A.  V132 Wirth L.  V91 Wirth M.  V879 Wirths S.  P444, V662, V861 Wirtz R.M.  P461 Wischmann V.  P203 Wisplinghoff H.  P797, P798 Witham D.  V857 Witte J.  P790 Wittig S.  V427 Wittke G.  P505 Wittke K.  P193 Witzens-Harig M.  P228, V74, V546, V655, V659, V863 Wlodarski M.  V607 Woessmann W.  V708 Wöhrer S.  V422 Wöhrl S.  P504, V752 Woike M.  P266, P267

Oncol Res Treat 2015;38(suppl 5):271–288

287


CONTENTS AUTHOR INDEX

Wolf A.  V656 Wolf D.  P246, P254, P754, P760, P813, V82, V161, V723 Wolf J.  P465, V44, V49, V884, V885, V887, V889 Wolf S.  V687 Wolf T.  V26, V657 Wolff A.  P205 Wolff D.  P489, V692, V860 Wolff H.  V733 Wolff T.  P256, P458 Wölfler A.  P245, P443, P453, P815, V307 Wolfram M.  P822 Wolfrum P.  V752 Wöll E.  P453, V314, V694 Wollina K.  P491 Wolschke C.  V627 Wolters M.  V627 Wömpner C.  V887 Wood K.  V673 Worden F.P.  V93 Wörtz I.  P509 Wroblewski M.  P773 Wuchter P.  P186, P243, V125, V546, V863 Wulf G.G.  V305, V425 Wurm A.  V541 Wyen C.  V657 Wyler S.F.  V369 X Xu L.  P211

288

Y Yan Y.  V671 Ychou M.  P211 Yin S.  P824 Yoshino T.  P211 Yovine A.  V889 Yu P.  P474 Yu Y.  V28 Yue B.  V28

Z Zackova D.  V603 Zadrozny N.  P233 Zagrijtschuk O.  P759 Zaiss M.  P235, P255 Zamora P.  P799 Zanchini R.  P457 Zanivan S.  V310 Zapfel S.  P485 Zaritskey A.  V603 Zaum M.  P271 Zaun S.  V393 Zaunig S.  V706 Zebisch A.  P245, P443, P815, V307 Zeck S.  V549 Zeilhofer H.-F.  V615 Zeis M.  V40 Zeiser R.  V161, V420, V698, V858 Zelenetz A.D.  V293 Zeller A.  V34 Zenhäusern R.  P260 Zentner A.  P512 Zenz T.  V294, V420

Oncol Res Treat 2015;38(suppl 5):272–288

Zeremski V.  P508, V707 Zettl F.  V377 Zeus T.  V753 Zhan L.  P265 Zhang L.  V73, V559 Zhang Z.  V341 Zhu J.  V91 Ziegenhain C.  V343 Ziegler R.  V714 Zielinski C.C.  P499 Ziepert M.  V656 Zilkens C.  P174 Zimber J.  V682 Zimmermann Y.  P216, P221, P223 Zimon D.  P502, V631 Zinngrebe B.  V657 Zintl P.  P227 Zipfel M.  P258 Zips D.  V583 Zober A.  P776 Zoellner A.-K.  P221, P223, V127 Zollikofer C.  P469 Zolnierek J.  V673 Zonder J.  V548 Zorn M.  V40 Zuber J.  P755, V621 Zucca E.  V584 Zuffa E.  P457 Zugelder L.  P786 Zugmaier G.  P175, V134 Zur Hausen G.  P214, P827 Zwick C.  V656 Zwierzina H.  P500

Author Index


CONTENTS AUTHOR INDEX

Imprint Oncol Res Treat 2015;38(suppl 5):290

ISSN Print Edition: 2296–5270 ISSN Online Edition: 2296–5262 Journal Homepage: www.karger.com/ort Publication Data: Volume 38, 2015 of ‘Oncology Research and Treatment’ appears with 12 issues. Copyright: © 2015 by S. Karger Verlag für Medizin und Naturwissenschaften GmbH, Freiburg (Germany). All rights reserved. No part of the journal may be reproduced in any form without the written permission of the publisher. This includes digitalisation and any further electronic computing, like saving, copying, printing or electronic transmission of digitalized material from this journal (online or offline). Authorization to photocopy items for internal or personal use of specific clients is granted by Karger. Photocopying: This journal has been registered with the Copyright Clearance Center (CCC), as indicated by the code appearing on the first page of each ­article. For readers in the US, this code signals consent for copying of articles for personal or internal use, or for the personal or internal use of specific c­ lients, provided that the stated fee is paid per copy directly to Copyright Clearance Center Inc., 222 Rosewood Drive, Danvers, MA 01923 (USA) A copy of the first page of the article must accompany payment. Consent does not extend to copying for general distribution, for promotion, for creating new works, or for resale. In these cases, specific written permission must be obtained from the copyright owner, S. Karger GmbH, Wilhelmstraße 20A, 79098 Freiburg (Germany). Disclaimer: The statements and data contained in this publication are solely those of the individual authors and contributors and not of the publisher and the editor(s). The appearance of advertisements in the journal is not a ­warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting form any ideas, methods, instructions or products referred to in the content or advertisements. Distribution and Subscription: Karger offers three types of subscription: Print Only, Online Only and the combined Print + Online. The basic annual subscription rate is the same for all three delivery forms; however, a fee for the combined print and online subscription is levied, and there is a postage and handling charge for Print Only and Print + Online. Subscriptions run for a full calendar year. Prices are given per volume.

© 2015 S. Karger GmbH, Freiburg Fax +49 761 4 52 07 14 Information@Karger.com www.karger.com

Accessible online at: www.karger.com/ort

Print subscription: EUR 261.– + postage and handling. Online subscription: EUR 261.–. Combined (print + online) subscription: 311.– + postage and handling. For customers in Germany: Please turn to your bookshop or to S. Karger Verlag für Medizin und Naturwissenschaften GmbH Wilhelmstr. 20A, 79098 Freiburg (Germany) Tel. +49 761 45 20 70, Fax +49 761 45 20 714 E-mail information@karger.com For customers in all other countries: Please contact your bookshop or S. Karger AG Allschwilerstr. 10, 4009 Basel (Switzerland) Tel. +41 61 3 06 11 11, Fax +41 61 3 06 12 34 E-mail karger@karger.com Advertising: Correspondence should be addressed to the publisher. S. Karger Verlag für Medizin und Naturwissenschaften GmbH Attn. Ellen Zimmermann (Head of Marketing) E-mail e.zimmermann@karger.com Price list No. 25 of January 1, 2015 is effective. V.i.S.d.P. (Person responsible according to the German Press Law): Sibylle Gross Type setting and printing: Kraft Druck GmbH, 76275 Ettlingen, Germany Supplement 5/2015 ISBN 978-3-318-05626-6 e-ISBN 978-3-318-05627-3


Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.