AE&M 62-6

Page 1

ISSN 2359-3997

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM Vol. 62 – No. 06 – December 2018

Archives of Endocrinology

and Metabolism

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

Archives of

Endocrinology

and Metabolism OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM


OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM Vol. 62 – No. 06 – December 2018

editorials 571 The challenge continues Marcello D. Bronstein

Archives of Endocrinology 572 Critical analysis of molecular tests in indeterminate thyroid nodules

OFFICIAL JOURNAL OF THE BRAZILIAN review 576 Can current molecular tests help in the diagnosis of indeterminate thyroid nodule FNAB? SOCIETY OF Carolina Ferraz ENDOCRINOLOGY original articles AND METABOLISM 585 The characteristics of blood glucose fluctuations in patients with fulminant type 1 diabetes mellitus in the stable stage Debora L. S. Danilovic, Suemi Marui

and Metabolism

Jie Wang, Bing-Li Liu, Zheng Li, Hui-Qin Li, Rui Sun, Yun Hu, Kok-Onn Lee, Lei Ye, Xiao-Fei Su, Jian-Hua Ma

591 Heart rate response to graded exercise test of elderly subjects in different ranges of TSH levels Rafael Cavalcante Carvalho, Patrícia dos Santos Vigário, Dhiãnah Santini de Oliveira Chachamovitz, Diego Henrique da Silva Silvestre, Pablo Rodrigo de Oliveira Silva, Mario Vaisman, Patrícia de Fátima dos Santos Teixeira

597 Risk factors for new-onset diabetes mellitus after kidney transplantation (NODAT): a Brazilian single center study Camila Lima, Amanda Grden, Thelma Skare, Paulo Jaworski, Renato Nisihara

602 Higher prevalence of permanent congenital hypothyroidism in the Southwest of Iran mostly caused by dyshormonogenesis: a five-year follow-up study Majid Aminzadeh

Archives of

609 Conversion to Graves disease from Hashimoto thyroiditis: a study of 24 patients

Beatriz Gonzalez-Aguilera, Daniela Betea, Laurence Lutteri, Etienne Cavalier, Vincent Geenen, Albert Beckers, Hernan Valdes-Socin

615 Sex effects on the association between sarcopenia EWGSOP and osteoporosis in outpatient older adults: data from the SARCOS study

Endocrinology

Alberto Frisoli Jr., Fabíola Giannattasio Martin, Antonio Carlos de Camargo Carvalho, Jairo Borges, Angela T. Paes, Sheila Jean McNeill Ingham

623 A pioneering RET genetic screening study in the State of Ceará, Brazil, evaluating patients with medullary thyroid cancer and at-risk relatives: experience with 247 individuals Maria Cecília Martins-Costa, Susan C. Lindsey, Lucas L. Cunha, Fernando Porto Carreiro-Filho, André P. Cortez, Marcelo E. Holanda, J. Wilson M. de Farias, Sérgio B. Lima, Luís A. Albano Ferreira, Pedro Collares Maia Filho, Cléber P. Camacho, Gilberto K. Furuzawa, Ilda S. Kunii, Magnus R. Dias-da-Silva, João R. M. Martins, Rui M. B. Maciel

and Metabolism

636 Rare complications of multikinase inhibitor treatment

Fabián Pitoia, Angélica Schmidt, Fernanda Bueno, Erika Abelleira, Fernando Jerkovich

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM 641 Selective screening for thyroid dysfunction in pregnant women: How often do low-risk women cease to be treated following

brief report

the new guidelines of the American Thyroid Association? Pedro Weslley Rosario

case reports 644 Early development of a gonadal tumor in a patient with mixed gonadal dysgenesis Sarah Crestian Cunha, Juliana Gabriel Ribeiro de Andrade, Camila Matsunaga de Angelis, Athanase Billis, Joaquim Murray Bustorff-Silva, Andréa Trevas Maciel-Guerra, Márcio Lopes Miranda, Gil Guerra-Júnior

648 Suprasellar chordoid glioma: a report of two cases Karina Danilowicz, Santiago Gonzalez Abbati, Soledad Sosa, Florencia Lustig Witis, Gustavo Sevlever

655 Hypothyroidism associated with short bowel syndrome in children: a report of six cases Ananda Castro Vieira Passos, Fábio de Barros, Durval Damiani, Beatriz Semer, Wendy Cira Justiniano Cespedes, Bruna Sannicola, Ana Cristina Aoun Tannuri, Uenis Tannuri


AND METABOLISM OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

Financial and editorial assistant: Roselaine Monteiro roselaine@endocrino.org.br Rua Botucatu, 572 – conjunto 83 – 04023-062 – São Paulo, SP Telefax: (11) 5575-0311 / 5082-4788

Archives of Endocrinology Online submission / Electronic publishing www.aem-sbem.com • www.scielo.br/abem

and Metabolism

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

Archives of

Rua Anseriz, 27, Campo Belo 04618-050 – São Paulo, SP. Fone: 11 3093-3300 www.segmentofarma.com.br • segmentofarma@segmentofarma.com.br

Publication code: 22884.12.18

Endocrinology

Informations: Title: Eternos caminhantes, 1919 Year: 1973 Author: Lasar Segall Technique: Oil on canvas OFFICIAL JOURNAL OF THE BRAZILIAN Measures: 138 x 184 cm SOCIETY OF ENDOCRINOLOGY AND METABOLISM Collection: Museum Lasar Segall-IBRAM/MinC

and Metabolism

All adds have to comply with RDC no 96/08 Commercial advising: Estela Kater estela.kater@gmail·com

Circulation of this issue: 3,500 copies Subscription: R$450.00/year – Single issue: R$55.00 Indexed in Biological Abstracts, Index Medicus, Latindex, Lilacs, MedLine, SciELO, Scopus, ISI-Web of Science BRAZILIAN ARCHIVES OF ENDOCRINOLOGY AND METABOLISM Brazilian Society of Endocrinology and Metabolism – São Paulo, SP: Brazilian Society of Endocrinology and Metabolism, volume 5, 1955Six issues/year Continued from: Brazilian Archives of Endocrinology (v. 1-4), 1951-1955 ISSN 2359-3997 (printed issues) ISSN 2359-4292 (online issues) 1. Endocrinology – journals 2. Metabolism – journals I. Brazilian Society of Endocrinology and Metabolism II. Brazilian Medical Association CDU 612.43 Endocrinology CDU 612.015.3 Metabolism

Supported by:


OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

OFFICIAL JOURNAL OF THE BRAZILIAN SOCIETY OF ENDOCRINOLOGY AND METABOLISM

Archives of endocrinology and metabolism Official journal of SBEM – Brazilian Society of Endocrinology and Metabolism (Department of the Brazilian Medical Association), SBD – Brazilian Diabetes Society, ABESO – Brazilian Association for the Study of Obesity and Metabolic Syndrome

2015-2018 EDITOR-IN-CHIEF Marcello D. Bronstein (SP)

CO-EDITORS

REPRESENTATIVES OF COLLABORATING SOCIETIES SBD

Larissa Gomes (SP)

ABESO

Léa Maria Zanini Maciel (SP)

OFFICIAL JOURNALLeandro Kasuki (SP) OF THE BRAZILIANMadson Queiroz Almeida (SP) Brazilian Editorial Commission Manoel Ricardo Alves Martins (CE) Alexander A. L. Jorge (SP) OF SOCIETY Marcio Mancini (SP) Alexandre Hohl (SC) ENDOCRINOLOGY Margaret Cristina S. Boguszewski (PR) Ana Amélia Hoff (SP) AND METABOLISM Maria Candida B. V. Fragoso (SP) Ana Claudia Latronico (SP) Maria Edna de Melo (SP)

and Metabolism

Ana Luiza Silva Maia (RS)

Maria Izabel Chiamolera (SP)

André Fernandes Reis (SP)

Maria Marta Sarquis (MG)

Andrea Glezer (SP)

Mario Saad (SP)

Tânia S. Bachega (SP)

Antônio Marcondes Lerário (SP)

Mário Vaisman (RJ)

INTERNATIONAL ASSOCIATE EDITOR

Antônio Roberto Chacra (SP)

Marise Lazaretti Castro (SP)

Ayrton Custódio Moreira (SP)

Milena Caldato (PA)

Shlomo Melmed (Los Angeles, EUA)

Berenice B. Mendonça (SP)

Raquel Soares Jallad (SP)

Bruno Halpern (SP)

Rodrigo Moreira (RJ)

Carlos Alberto Longui (SP)

Ruth Clapauch (RJ)

Archives of

FOUNDER

ASSOCIATE EDITORS

Waldemar Berardinelli (RJ)

PRESIDENTS OF THE SBEM DEPARTMENTS

EDITORS-IN-CHIEF, EDITORIAL OFFICE*

ADRENAL AND HYPERTENSION

Endocrinology César Luiz Boguszewski (PR)

Sandra R. G. Ferreira (SP)

Clarisse Ponte (CE)

Simão A. Lottemberg (SP)

Delmar Muniz Lourenço Jr. (SP)

Sonir Roberto Antonini (SP)

Luiz Alberto Andreotti Turatti (SP)

Denise Momesso (RJ)

Suemi Marui (SP)

DYSLIPIDEMIA AND ATHEROSCLEROSIS

Eder Carlos R. Quintão (SP)

Madson Queiroz de Almeida (SP)

1951-1955 Waldemar Berardinelli (RJ) Thales Martins (RJ)

DIABETES MELLITUS

1957-1972 Clementino Fraga Filho (RJ)

Cynthia Melissa Valério (RJ)

Edna Nakandakare (SP)

1964-1966* Luiz Carlos Lobo (RJ)

BASIC ENDOCRINOLOGY

Edna T. Kimura (SP)

Maria Izabel Chiamolera (SP)

1966-1968* Pedro Collett-Solberg (RJ) 1969-1972* João Gabriel H. Cordeiro (RJ)

and Metabolism

Weiss (RJ)

PEDIATRIC ENDOCRINOLOGY

Julienne Ângela Ramires de Carvalho (PR)

1983-1990 Antônio Roberto Chacra (SP)

BONE AND MINERAL METABOLISM

1995-2006 Claudio Elias Kater (SP) 2007-2010 Edna T. Kimura (SP) 2011-2014 Sergio Atala Dib (SP)

Elaine Maria Frade Costa (SP)

OFFICIAL JOURNAL OF THE FelipeBRAZILIAN Gaia (SP) SOCIETY OF ENDOCRINOLOGY AND METABOLISM Rita de Cássia Viana Vasconcellos Flavio Hojaij (SP) FEMININE ENDOCRINOLOGY AND ANDROLOGY

1978-1982 Armando de Aguiar Pupo (SP)

1991-1994 Rui M. de Barros Maciel (SP)

Laércio Joel Franco (SP)

Luiz Alberto Andreotti Turatti (SP)

Archives of Endocrinology Bruno Ferraz de Souza (SP) Erika Parente (SP) Francisco Bandeira (PE) Fernanda Vaisman (RJ) Fernando M. A. Giuffrida (BA) João Roberto Maciel Martins (SP) Melanie Rodacki (RJ) Monica R. Gadelha (RJ) Nina Rosa C. Musolino (SP) Poli Mara Spritzer (RS) Ricardo Meirelles (RJ) Rogerio Friedman (RS) Rui M. B. Maciel (SP)

Julio Z. Abucham (SP)

Carolina Aguiar Moreira (PR) NEUROENDOCRINOLOGY

Marcello Delano Bronstein (SP) OBESITY

Maria Edna de Melo (SP) THYROID

Célia Regina Nogueira (SP)

Victória Borba (PR)

International Editorial Commission Andrea Giustina (Itália)

Gil Guerra-Júnior (SP)

Antonio C. Bianco (EUA)

Giovanna Balarini Lima (RJ)

Décio Eizirik (Bélgica)

Gisah M. do Amaral (SP)

Franco Mantero (Itália)

Hans Graf (SP)

Fernando Cassorla (Chile)

José Augusto Sgarbi (SP)

Gilberto Paz-Filho (Austrália)

José Gilberto H. Vieira (SP)

John P. Bilezikian (EUA)


SBEM – Brazilian Society of Endocrinology and Metabolism SBEM BRAZILIAN BOARD OF DIRECTORS 2017-2018 President Vice-President Executive Secretary Adjunct Executive Secretary Treasurer-General Adjunct Treasurer

Fábio Rogério Trujilho Alexandre Hohl Paulo Augusto Carvalho de Miranda Neuton Dornelas Gomes Rodrigo de Oliveira Moreira Marcio Corrêa Mancini

Rua Humaitá, 85, cj. 501 22261-000 – Rio de Janeiro, RJ Fone/Fax: (21) 2579-0312/2266-0170 www.endocrino.org.br sbem@endocrino.org.br

Scientific Departments - 2017/2018 Brazilian Society of Endocrinology and Metabolism ADRENAL AND HYPERTENSION

DIABETES MELLITUS

President Madson Queiroz de Almeida madsonalmeida@usp.br

President Luiz Alberto Andreotti Turatti turatti@uol.com.br Directors Amely Pereira Silva Balthazar Gustavo José Caldas Pinto Costa Sergio Alberto Cunha Vêncio Walter José Minicucci Thaísa Dourado Guedes Treasurer João Eduardo Nunes Salles Alternates Marcos Cauduro Troian Victor Gervásio e Silva

Vice-President Directors

Marivânia da Costa Santos Alexis Dourado Guedes Flávia Amanda Costa Barbosa Milena Coelho Fernandes Caldato Sonir Roberto Rauber Antonini Tânia Aparecida Sanchez Bachega

DYSLIPIDEMIA AND ATHEROSCLEROSIS

BASIC ENDOCRINOLOGY

President Cynthia Melissa Valério cy_valerio@yahoo.com.br

President Maria Izabel Chiamolera mchiamolera@unifesp.br Vice-President Bruno Ferraz de Souza Directors Catarina Segreti Porto Dóris Rosenthal Maria Tereza Nunes Marisa Maria Dreyer Breitenbach Tania Maria Ruffoni Ortiga Alternates Vânia Maria Corrêa da Costa Ubiratan Fabres Machado

Vice-President Directors

Renan Magalhães Montenegro Júnior Fernando de Mello Almada Giuffrida Marcello Casaccia Bertoluci


Scientific Departments - 2017/2018 WOMEN ENDOCRINOLOGY AND ANDROLOGY President Rita de Cássia Viana Vasconcellos Weiss rcvweiss@gmail.com Vice-President Directors Alternates

Dolores Perovano Pardini Amanda Valéria Luna de Athayde Mônica de Oliveira Poli Mara Spritzer Ricardo Martins da Rocha Meirelles Ruth Clapauch Izydorczyk Antônio Mendes Fontanelli Larissa Garcia Gomes

PEDIATRIC ENDOCRINOLOGY President Julienne Angela Ramires de Carvalho julienne@endocrinoped.com.br Vice-President Directors Alternate

Carlos Alberto Longui Aline da Mota Rocha Angela Maria Spinola e Castro Cláudia Braga Monteiro Paulo César Alves da Silva Suzana Nesi França Marilia Martins Guimarães

BONE AND MINERAL METABOLISM

NEUROENDOCRINOLOGY

President Vice-President Directors Alternate

President Marcello D. Bronstein mdbronstein@uol.com.br Vice-President César Luiz Boguszewski Directors Heraldo Mendes Garmes Luciana Ansanelli Naves Lucio Vilar Rabelo Filho Luiz Antonio de Araujo Mônica Roberto Gadelha Alternates Andrea Glezer Manoel Ricardo Alves Martins

Carolina Aguiar Moreira carolina.aguiar.moreira@gmail.com Miguel Madeira Barbara Campolina Carvalho Silva Francisco Alfredo Bandeira e Farias Marise Lazaretti Castro Sergio Setsuo Maeda Victória Zeghbi Cochenski Borba Tatiana Munhoz da Rocha Lemos Costa

OBESITY

THYROID

President Maria Edna de Melo medna@usp.br Vice-President Rosana Bento Radominski Director/Secretary Walmir Ferreira Coutinho Director/Treasurer Erika Paniago Guedes Directors Cintia Cercato Leila Maria Batista Araujo Director/Treasurer Fabio Ferreira de Moura Alternate Jacqueline Rizzolli

President Célia Regina Nogueira nogueira@fmb.unesp.br Vice-President José Augusto Sgarbi Secretary Janete Maria Cerutti Directors Ana Luiza Silva Maia Laura Sterian Ward Patricia de Fátima dos Santos Teixeira Gisah Amaral de Carvalho Mario Vaisman Alternate Danilo Glauco Pereira Villagelin Neto


Permanent Commissions - 2017/2018 Brazilian Society of Endocrinology and Metabolism STRATEGIC PLANNING FOLLOW-UP

HISTORY OF ENDOCRINOLOGY

President Alexandre Hohl alexandrehohl@uol.com.br Members Nina Rosa de Castro Musolino, Airton Golbert, Ricardo Martins da Rocha Meirelles, Ruy Lyra da Silva Filho

President Henrique de Lacerda Suplicy hsuplicy@gmail.com Members Adriana Costa e Forti, Thomaz Rodrigues Porto da Cruz

ENDOCRINOLOGY CAMPAIGNS

President

César Luiz Boguszewski

President Erika Bezerra Parente ebparente@gmail.com Members Érika Paniago Guedes, Teresa Arruti Rey

cesarluiz@hc.ufpr.br

Members

Ruy Lyra da Silva Filho, Valéria Cunha C. Guimarães, Ana Cláudia Latrônico

SCIENTIFIC COMISSION President Alexandre Hohl alexandrehohl@uol.com.br Indicated by the directories Joao Eduardo Nunes Salles, Alexis Dourado Guedes, Erika Bezerra Parente, Ana Mayra Andrade de Oliveira, Margaret Cristina da Silva Boguszewski, Guilherme Alcides Flores Rollin, Milena Coelho Fernandes Caldato, Mônica de Oliveira, Nina Rosa de Castro Musolino

SOCIAL COMMUNICATION President Ricardo Martins da Rocha Meirelles r.meirelles@terra.com.br Nominated by the president Alexandre Hohl ABEM Editor Marcello D. Bronstein Members Nina Rosa de Castro Musolino

CONTINUOUS MEDICAL EDUCATION President Alexandre Hohl alexandrehohl@uol.com.br Members Lireda Meneses Silva, Walter José Minicucci, Cleber Favaro

STATUTES, RULES AND REGULATIONS President Nina Rosa de Castro Musolino ninamusolino@gmail.com Members Airton Golbert, Henrique de Lacerda Suplicy, Luiz Henrique Maciel Griz Representative of the Evandro de Souza Portes brazilian directory

PROFESSIONAL ETHICS AND DEFENCE President Itairan da Silva Terres itairan.terres@gmail.com Vice-Inspector Maite Trojaner Salona Chimeno 1ST member Diana Viegas Martins 2ND member João Modesto Filho 3RD member Evandro de Souza Portes 4TH member Marcelo Henrique da Silva Canto Costa 5TH member Luiz Henrique Santos Canani

ENDOCRINE DYSREGULATORS President Elaine Frade Costa elainefradecosta@gmail.com Vice-President Margaret Cristina da Silva Boguszewski Members Tania Aparecida Sanchez Bachega, Ricardo Martins da Rocha Meirelles, Marilia Martins Guimarães, Eveline Gadelha Pereira Fontenele, Maria Izabel Chiamolera

INTERNATIONAL

NORMS, QUALIFICATION AND CERTIFICATION President Vivian Carole Moema Ellinger vivianellinger@gmail.com Members Ronaldo Rocha Sinay Neves, Marisa Helena César Coral, Maria Emilia Pereira de Almeida, Milena Coelho Fernandes Caldato

JOINT COMMISSION – CAAEP President Julienne Angela Ramires de Carvalho julienne@endocrinoped.com.br Members Marilia Martins Guimarães, Suzana Nesi França

RESEARCH President Members

Freddy Eliaschewitz freddy.g@uol.com.br Antônio Roberto Chacra, Luiz Augusto Tavares Russo

GUIDELINES PROJECT Coordinator Alexis Dourado Guedes dr.alexis@uol.com.br Adrenal and hypertension Madson Queiroz de Almeida Dyslipidemia and atherosclerosis Cynthia Melissa Valério Diabetes Mellitus Luiz Alberto Andreotti Turatti Basic endocrinology Maria Izabel Chiamolera Feminine and Andrology Rita de Cássia Viana Vasconcellos Weiss Pediatric Endocrinology Julienne Angela Ramires de Carvalho Bone and mineral metabolism Carolina Aguiar Moreira Neuroendocrinology Marcello D. Bronstein Obesity Maria Edna de Melo Thyroid Célia Regina Nogueira

TEMPORARY – SPORT AND EXERCISE ENDOCRINOLOGY - CTEEE President: Yuri Galeno Pinheiro Chaves de Freitas yurigaleno@gmail.com Members: Fábio Ferreira de Moura, Clayton Luiz Dornelles Macedo, Roberto Luís Zagury, Ricardo de Andrade Oliveira, Fulvio Clemo Thomazelli, Felipe Henning Duarte Gaia

TITLE OF SPECIALIST IN ENDOCRINOLOGY AND METABOLISM President: Josivan Gomes de Lima josivanlima@gmail.com Vice-President: Márcio Corrêa Mancini Members: Marise Lazaretti Castro, Mauro Antônio Czepielewski, Milena Coelho Fernandes Caldato, Renan Magalhães Montenegro Júnior, Rogério Friedman

VALORIZATION OF NEW LEADERSHIPS President Joaquim Custodio da Silva Junior jocsjunior@uol.com.br Members Joaquim Custodio da Silva Junior, Eduardo Quadros Araújo, Marcelo Fernando Ronsoni, Manoel Ricardo Alves Martins, Marcio Weissheimer Lauria


Brazilian Societies and Associations for Endocrinology and Metabolism

SBD – BRAZILIAN DIABETES SOCIETY SBD BRAZILIAN BOARD OF DIRECTORS (2018/2019)

President

Hermelinda Cordeiro Pedrosa

Vice-Presidents

Gustavo Caldas

Janice Sepulveda Reis

João Eduardo Nunes Salles

Rosane Kupfer

Rosângela Réa

1ST Secretary

Karla Melo

2ND Secretary

Fernanda Thomé

1ST Treasurer

Antonio Carlos Lerário

2ND Treasurer

Luiz Antônio Araújo

Supervisory Board

Silmara Leite

Regina Calsolari

Nely Calegaro

Estela Muskat Jatene

Rua Afonso Brás, 579, cj. 72/74 04511-011– São Paulo, SP Fone/Fax: (11) 3842-4931 secretaria@diabetes.org.br www.diabetes.org.br Administrative Manager: Anna Maria Ferreira

ABESO – BRAZILIAN ASSOCIATION FOR THE STUDY OF OBESITY AND METABOLIC SYNDROME ABESO BRAZILIAN BOARD OF DIRECTORS (2017-2018)

President

Maria Edna de Melo

Vice-President

Alexander Koglin Benchimol

1ST Secretary General

Bruno Halpern

2ND Secretary General

Fábio Ferreira de Moura

Treasurer

Erika Paniago Guedes

Rua Mato Grosso, 306, cj. 1711 01239-040 – São Paulo, SP Fone: (11) 3079-2298/Fax: (11) 3079-1732 Secretary: Renata Felix info@abeso.org.br www.abeso.org.br



editorial

The challenge continues Marcello D. Bronstein1

F

our years have elapsed since I took the post of Editor-in-Chief and changed the traditional “Arquivos Brasileiros de Endocrinologia e Metabologia” to “Archives of Endocrinology and Metabolism” (AE&M). This modification didn’t affect the pertinence of the journal to Brazil (it remained the official journal of the Brazilian Society of Endocrinology and Metabolism) but, using English as the official language, became more available to the scientific word. As a matter of fact, the submission of articles from abroad increased substantially, ranging from 44.8% to 52.3% of total, encompassing about 50 different countries, despite the still bigger participation of Brazilian papers. Among the submissions from abroad, the top ten countries were Turkey, China, Iran, Portugal, India, Argentina, USA, Italy, Mexico and Greece, in decrescent order. The amount of total submissions of manuscripts per year (including Brazil) was 306 in 2015, 337 in 2016 and 320 in 2017 (2018 is still ongoing), with an acceptance rate of about 25% of total submissions. Concerning the impact factor, the marker of a Journal’s relevance, it increased from 0.056 as the last “Arquivos Brasileiros de Endocrinologia e Metabologia” to 1.076 – the first impact factor of AE&M. This achievement could not be attained without the dedicated and competent collaboration of our Associated Editors, as well as the Editorial Board and all the reviewers. I wish to express a heartfully acknowledgement and gratitude to all of them. I was honored by my pairs with an additional four years as Editor-in-Chief of the AE&M. I thank them for their confidence and will do my best to accomplish the task to improve our Journal’s relevance in the scientific community.

Editor-chefe da Archives of Endocrinology and Metabolism, chefe da Unidade de Neuroendocrinologia, Serviço de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil 1

Correspondence to: Marcello D. Bronstein aem.editorial.office@endocrino.org.br Received on Nov/20/2018 Accepted on Nov/20/2018 DOI: 10.20945/2359-3997000000094

Copyright© AE&M all rights reserved.

Disclosure: no potential conflict of interest relevant to this article was reported.

Arch Endocrinol Metab. 2018;62/6

571


editorial

Critical analysis of molecular tests in indeterminate thyroid nodules Debora L. S. Danilovic1,2, Suemi Marui1

Laboratório de Endocrinologia Celular e Molecular (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil 2 Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil 1

Correspondence to: Debora L. S. Danilovic Laboratório de Endocrinologia Celular e Molecular (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo Av. Dr. Arnaldo, 455, sala 4305 01246-903 – São Paulo, SP, Brasil deboradanilovic@usp.br Received on Nov/23/2018 Accepted on Nov/23/2018

Copyright© AE&M all rights reserved.

DOI: 10.20945/2359-3997000000095

572

T

he prevalence of thyroid nodules has increased in the last decades, mostly due to the widespread use of cervical imaging for investigation of pathologies not related to thyroid (incidentalomas) and, eventually, thyroid imaging screening for individuals at no risk for thyroid disease. Nevertheless, thyroid carcinomas correspond to 5%15% of thyroid nodules and the failure in identifying benign nodules in asymptomatic patients usually leads to unnecessary thyroidectomies. Ultrasonography and fine needle aspiration biopsy (FNAB) contribute to preoperative diagnosis, but indeterminate cytology still represents 20% to 30% of diagnosis, namely Bethesda III, IV and V, with rates of malignancy reported as 10%-30%, 25%-40% and 50%-75%, respectively (1). In this context, the recent knowledge of molecular abnormalities related to thyroid cancer has been used to improve patient outcome, not only to avoid diagnostic surgeries and enable active surveillance, but also to guide the extension of thyroidectomy (total or partial), particularly in Bethesda categories III and IV nodules. In this issue, Ferraz C. reviewed molecular tests developed to improve the diagnosis of indeterminate biopsies (2). A classification according to their predominant ability to “rule-in” and/or “rule-out” cancer was proposed. She pointed out that a desirable test to predict malignancy would have high positive predictive value, while prediction of benign nodules would require high negative predictive values. It is not easy to critically analyze a molecular test to diagnose thyroid cancer. The performance of each molecular test should be based on its characteristics, given by sensitivity, specificity and predictive values. Sensitivity and specificity correspond to the rate of thyroid cancer and of benign nodules detected by the test. Positive predictive value (PPV) is the proportion of thyroid cancer among positive test results and negative predictive value (NPV) is the proportion of benign nodules (not cancer) among negative results, which are both dependent on the prevalence of cancer in the studied population. Therefore, if you consider the probability of thyroid cancer in Bethesda category III and IV around 25%, a good “rule-out” test would have sensitivity > 90% to obtain a NPV > 94%, and a good “rule-in” test would have specificity > 80% to result in a PPV > 60%. The first molecular test commercially available in Brazil was DNA sequencing to detect BRAF mutation. We prospectively evaluated the presence of the p.V600E mutation of the BRAF gene, and also searched for N-RAS, H-RAS, K-RAS mutations, in FNAB of Bethesda categories III and IV (3). BRAF mutation was detected in 65% of carcinomas included in our analysis. This simple test had specificity of 100% and PPV of 100% in both Bethesda categories III and IV. However, sensitivities were low, 35% and 57%, resulting in NPVs of 81% and 86%, respectively. When you order BRAF mutation test, a “positive” result assures 100% chance of malignancy but, if Arch Endocrinol Metab. 2018;62/6


“negative”, the nodule is still considered indeterminate and a diagnostic surgery is necessary. Performance of such “rule in” test was improved by additional evaluation of PAX8/PPARg, RET/ PTC1, RET/PTC 3 rearrangements, “7-gene panel” (4), currently available as ThyGenX®. Despite improvement, particularly in Bethesda category III, independent clinical validation studies did not replicate the performance, and a desirable NPV to avoid surgery was not reached, as false negative results would occur in more than 5% of the cases. As reviewed by Ferraz C., new technologies, especially the next generation sequencing (NGS), provided a significant step-forward to clinical acceptance of molecular tests in the preoperative evaluation of thyroid nodules (2). The Afirma® gene expression classifier (GEC) differentiates benign and malignant nodules based on patterns of mRNA expression (5). It was proved to be a cost-effective “rule-out” test to avoid surgery due to high sensitivity and NPV around 95%. As benign results were obtained in 41% of evaluated nodules in a clinical validation study, it has been suggested to be also cost-saving, since almost one out of two molecular tests would avoid one diagnostic surgery. Afirma® GEC has been extensively evaluated by different and independent centers. Similarly to sevengene panel, post-validation trials usually did not submit all patients to surgery, as a matter of fact, most of the “benign” GEC did not undergo surgical intervention, which could mislead an excellent performance. The cost-saving capacity was not confirmed, since depending on patients’ selection, more tests were necessary to avoid one surgery. Besides, it became evident that the performance of the test relied on the prevalence of malignancy of the studied population. If the rate of malignancy was lower than 25%, the cost-effectiveness of Afirma® GEC decreased, as fewer “suspicious” results corresponded to thyroid carcinomas. A novel Afirma® gene sequencing classifier (GSC) has been recently developed to improve evaluation of RNA expression and GSC increased specificity, particularly to recognize more Hürthle lesions as “benign”, preserving its high sensitivity (6). Some molecular tests were reported as being not only able to correctly identify most of thyroid carcinomas, but also most of the benign lesions called “rule-in and rule-out” tests. ThyroSeq® v2 with expanded panel of mutations, rearrangements and gene expressions, particularly in Bethesda category IV, is Arch Endocrinol Metab. 2018;62/6

apparently efficient in indicating surgery if “positive”, and to consider follow-up without diagnostic surgery if “negative” (7). Post-validation studies demonstrated the usefulness of ThyroSeq® v2 molecular test to avoid surgeries, as most of “negative” results were not submitted to surgical procedures, confirming it as a good “rule-out” test. However, as regards Bethesda category III lesions, Thyroseq® v2 presented poorer performance and did not prove to be such a good “rule-in and rule-out” test. More recently, Nikiforova and cols. developed a new version of ThyroSeq® v3, which provides a genomic classifier (GC) score calculated according to the strength of association of detected genetic alterations with malignancy (8). Since the presence of a mutated gene is not synonymous of malignancy, Thyroseq® v3 presents different reports for “negative” and “positive” results. There are two classes of “negative” results: “negative”, as expected to be a benign lesion and “currently negative”, when a mutation is found in a low-risk gene that by itself is not sufficient to full cancer development (i.e., mutation in PTEN, EIF1AX) or it is found in a subpopulation of cells. Although at the time of sampling most of these nodules are benign, some of them may undergo clonal expansion and acquire additional mutations, so active surveillance is suggested, considering to repeat FNA and, possibly, molecular testing after one year of followup. When test result is “positive”, the prognosis is promptly suggested: “low-risk”, when RAS-positive is found, and ‘high risk’, when TERT and BRAF-positive carcinoma are present. Therefore, multicenter clinical trials are necessary to validate its performance. Finally, microRNA (miRNA) gene expression classifiers have also been developed to improve diagnostic performance of Bethesda categories III and IV, ThyraMIR® combined with ThygenX® (9), Rosetta GX Reveal® (10) and the Brazilian mir-THYpe® (11). Their main limitation is lack of multicenter experience. MicroRNAs panels should be more extensively studied in order to confirm their performance as desirable both “rule-in and rule-out’” tests. A remark should be made about molecular tests in the recently proposed noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP), which corresponds to the noninvasive encapsulated follicular variant of papillary thyroid carcinoma. Molecular tests available nowadays proved to be unable to classify NIFTPs as benign lesions. In retrospective evaluations, 81% out of 32 NIFTPs analyzed by Afirma® 573

Copyright© AE&M all rights reserved.

Molecular diagnostic tests in thyroid nodules


Molecular diagnostic tests in thyroid nodules

GEC had “suspicious” results (12). Likewise, 3 out of 5 NIFTPs submitted to ThyroSeq® v.2 had “positive” results (13). Meanwhile, treatment of NIFTP is still surgical removal due to the potential risk of progression to invasive carcinoma. Therefore, “suspicious” or “positive” results in “rule-in and/or rule-out”’ test will not change NIFTP management.

Copyright© AE&M all rights reserved.

WHICH TEST SHOULD BE CHOSEN? Only a few institutions reported their experience and compared the performance of molecular tests in similar conditions. In one of them, two-thirds of Bethesda categories III and IV lesions were managed nonoperatively based on nonsuspicious results of Thyroseq® v2 or Afirma® GEC. Considering the rate of malignancy of 14%, Livhits and cols. (14) demonstrated that ThyroSeq® v.2 had a better performance to identify malignancy compared to GEC (PPV 57% vs. 39%). Similarly, Jug and cols. demonstrated that “negative” results in molecular tests helped to reduce surgery indication in ~50% of patients (15). Considering the rate of malignancy of only 12%, ThyroSeq® v.2 had PPV of 40% and 50% in Bethesda categories III and IV and GEC had PPV of 29% in Bethesda category III and 27% in Bethesda category IV. Therefore, the performance of the molecular tests must be carefully interpreted, considering that different populations, diverse prevalence of malignancy, and the fact that not all patients were submitted to confirmatory surgery altogether, interfere in results when compared to clinical validation studies. While microRNA panels have limited multicenter experience, we could suggest that both Afirma® GEC/ GSC and ThyroSeq® v2/v3 might be used to improve preoperative diagnosis of Bethesda categories III and IV lesions. High cost and no health insurance coverage limit the widespread application of molecular tests in Brazil and other countries. It is always important to consider risk factors, patient´s clinical conditions and desire, and, certainly, US characteristics before choosing a molecular test. We usually wish to identify benign lesions in order to defer diagnostic surgeries. High-risk nodules at US may not benefit from “rule-out” molecular test to avoid surgery. Actually, in high-risk nodules, a “positive” result in a “rule-in” test, reinforcing malignancy, is more useful, as a partial diagnostic surgery may turn into total thyroidectomy to treat cancer. On the other hand, if we evaluate an indeterminate or low-risk nodule at US, 574

“rule-out” tests seem more relevant because of their low rate of false-benign results. Implementation of molecular test into routine clinical practice should be made with cautious, as long-term outcome data on companion use of molecular test to guide therapeutic decision-making is currently lacking. To conclude, as The American Thyroid Association strongly recommends: “if molecular testing is being considered, patients should be counseled regarding the potential benefits and limitations of testing and about the possible uncertainties in the therapeutic and longterm clinical implications of results” (1). Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016;26(1):1-133. 2. Ferraz C. Can current molecular tests help in the diagnosis of indeterminate thyroid nodule FNAB?. Arch Endocrinol Metab. 2018;62(6):576-84. 3. Danilovic DL, Lima EU, Domingues RB, Brandao LG, Hoff AO, Marui S. Pre-operative role of BRAF in the guidance of the surgical approach and prognosis of differentiated thyroid carcinoma. Eur J Endocrinol. 2014;170(4):619-25. 4. Nikiforov YE, Steward DL, Robinson-Smith TM, Haugen BR, Klopper JP, Zhu Z, et al. Molecular testing for mutations in improving the fine-needle aspiration diagnosis of thyroid nodules. J Clin Endocrinol Metab. 2009;94(6):2092-8. 5. Chudova D, Wilde JI, Wang ET, Wang H, Rabbee N, Egidio CM, et al. Molecular classification of thyroid nodules using high-dimensionality genomic data. J Clin Endocrinol Metab. 2010;95(12):5296-304. 6. Patel KN, Angell TE, Babiarz J, Barth NM, Blevins T, Duh QY, et al. Performance of a Genomic Sequencing Classifier for the Preoperative Diagnosis of Cytologically Indeterminate Thyroid Nodules. JAMA Surg. 2018;153(9):817-24. 7. Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, et al. Highly accurate diagnosis of cancer in thyroid nodules with follicular neoplasm/suspicious for a follicular neoplasm cytology by ThyroSeq v2 next-generation sequencing assay. Cancer. 2014;120(23):3627-34. 8. Nikiforova MN, Mercurio S, Wald AI, Barbi de Moura M, Callenberg K, Santana-Santos L, et al. Analytical performance of the ThyroSeq v3 genomic classifier for cancer diagnosis in thyroid nodules. Cancer. 2018;124(8):1682-90. 9. Labourier E, Shifrin A, Busseniers AE, Lupo MA, Manganelli ML, Andruss B, et al. Molecular Testing for miRNA, mRNA, and DNA on Fine-Needle Aspiration Improves the Preoperative Diagnosis of Thyroid Nodules With Indeterminate Cytology. J Clin Endocrinol Metab. 2015;100(7):2743-50. 10. Lithwick-Yanai G, Dromi N, Shtabsky A, Morgenstern S, Strenov Y, Feinmesser M, et al. Multicentre validation of a microRNA-based Arch Endocrinol Metab. 2018;62/6


Molecular diagnostic tests in thyroid nodules

assay for diagnosing indeterminate thyroid nodules utilising fine needle aspirate smears. J Clin Pathol. 2017;70(6):500-7. 11. Santos MT, Buzolin AL, Gama RR, Silva ECA, Dufloth RM, Figueiredo DLA, et al. Molecular classification of thyroid nodules with indeterminate cytology: development and validation of a highly sensitive and specific new miRNA-based classifier test using fine-needle aspiration smear slides. Thyroid. 2018. doi: 10.1089/thy.2018.0254. [Epub ahead of print]

14. Livhits MJ, Kuo EJ, Leung AM, Rao J, Levin M, Douek ML, et al. Gene Expression Classifier vs Targeted Next-Generation Sequencing in the Management of Indeterminate Thyroid Nodules. J Clin Endocrinol Metab. 2018;103(6):2261-8. 15. Jug RC, Datto MB, Jiang XS. Molecular testing for indeterminate thyroid nodules: Performance of the Afirma gene expression classifier and ThyroSeq panel. Cancer Cytopathology. 2018;126:471-80.

CopyrightŠ AE&M all rights reserved.

12. Song SJ, LiVolsi VA, Montone K, Baloch Z. Pre-operative features of non-invasive follicular thyroid neoplasms with papillarylike nuclear features: An analysis of their cytological, Gene Expression Classifier and sonographic findings. Cytopathology. 2017;28(6):488-94.

13. Valderrabano P, Khazai L, Leon ME, Thompson ZJ, Ma Z, Chung CH, et al. Evaluation of ThyroSeq v2 performance in thyroid nodules with indeterminate cytology. Endocr Relat Cancer. 2017;24(3):127-36.

Arch Endocrinol Metab. 2018;62/6

575


review

Can current molecular tests help in the diagnosis of indeterminate thyroid nodule FNAB? Carolina Ferraz1

ABSTRACT Unidade de Doenças da Tireoide, Divisão de Endocrinologia, Departamento de Medicina, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, SP, Brasil 1

Correspondence to: Carolina Ferraz Av. Angélica, 2.491, cj 104 01232-011 – São Paulo, SP, Brasil carolina.ferraz.endocrinologia@gmail.com Received on Sept/25/2018 Accepted on Nov/6/2018 DOI: 10.20945/2359-3997000000081

Approximately 15–30% of all thyroid nodules evaluated with fine-needle aspiration biopsy (FNAB) are classified as cytologically indeterminate. The stepwise unraveling of the molecular etiology of thyroid nodules has provided the basis for a better understanding of indeterminate samples and an opportunity to decrease diagnostic surgery in this group of patients. Over the last 15 years, several studies have tested different methodologies to detect somatic mutations (by polymerase chain reaction and next-generation sequencing, for example), and to identify differentially expressed genes or microRNA, aiming at developing molecular tests to improve the presurgical diagnosis of cytologically indeterminate nodules. In this review, we will provide an overview of the currently available molecular tests and the impact of mutation testing on the diagnosis of thyroid cancer. We will also review current published data and future perspectives in molecular testing of thyroid nodule FNAB and describe the current Brazilian experience with this diagnostic approach. Based on currently available data, especially for countries outside the US-Europe axis, a rational use of these tests must be made to avoid errors with regard to test indication and interpretation of test outcomes. In addition to clinical, radiological, and cytological features, we still need to determine local malignancy rates and conduct more independent validation and comparative performance studies of these tests before including them into our routine approach to indeterminate FNAB. Arch Endocrinol Metab. 2018;62(6):576-84 Keywords Thyroid; nodules; molecular test; indeterminate samples

INTRODUCTION

Copyright© AE&M all rights reserved.

E

ven though the introduction of fine-needle aspiration biopsy (FNAB) has improved the selection of suspicious nodules for surgery (1), approximately 15% to 30% of all thyroid nodules undergoing FNAB are classified as cytologically indeterminate, which includes lesions of undetermined significance/follicular lesion of undetermined significance (AUS/FLUS; Bethesda III) or follicular neoplasm/suspicious for follicular neoplasm (FN/ SFN; Bethesda IV) (2,3). The stepwise unraveling of the molecular etiology of thyroid nodules has provided the basis for a better understanding of cytologically indeterminate nodules and a chance to reduce diagnostic surgery in this scenario. Because immunocytological markers have failed to show enough specificity and sensitivity, improved molecular testing for common somatic mutations (i.e., BRAF and RAS point mutations, or RET/PTC and PAX8/ PPARg rearrangements) and identification of gene and 576

microRNA (miRs) expression classifiers have emerged as the most promising approaches. During the last 15 years, several studies have tested different diagnostic methodologies to characterize thyroid cancer. The latest studies in this field have sought specific molecular markers to differentiate benign and malignant neoplasms and discriminate with increased sensitivity and specificity the different histotypes of thyroid cancer. In this review, we will provide a critical overview of the current impact of mutation testing on the diagnosis of thyroid cancer, discussing current possibilities and future perspectives in molecular testing of thyroid nodule FNABs. Additionally, we will describe the current Brazilian experience with molecular testing of thyroid nodules deemed indeterminate on FNABs.

Molecular testing from the beginning until now After serum-based biomarkers like calcitonin and thyroglobulin were first described, they provided Arch Endocrinol Metab. 2018;62/6


Molecular tests for indeterminate FNAB samples

mutually exclusive events, meaning, only one of these mutations is found in any particular cancer (14). When these mutations are used as independent biomarkers, their sensitivity and specificity are too low to be clinically relevant. However, a panel of mutations has been shown to improve both sensitivity and specificity rates (15). Nikiforov and cols. were the first group to report a gain in sensitivity (from 44% to 80%) and accuracy (from 93.3% to 97.4%) by analyzing a panel of BRAF, RAS, RET/PTC, and PAX8/PPARg mutations (15,16). Based on this evidence, the first commercially available test named “miRInform” (Asuragen, Inc., Austin, TX, USA) (Figure 1) was created in 2009. BRAF, KRAS, HRAS, NRAS, and chromosomal translocations resulting in RET/PTC1, RET/PTC3, and PAX8/PPARg fusions resulted in the development of a “7-Gene Panel.” This test was later replaced and is currently offered by Interpace Diagnostics (Parsippany, NJ, USA) as the ThyGenX test (Figure 1), with PIK3CA added to the panel. Next-generation sequencing (NGS) technologies or massively parallel sequencing are related terms that describe a DNA sequencing technology that has revolutionized genomic research and emerged as a

Copyright© AE&M all rights reserved.

the impetus for future research in the discovery of biomarkers for the diagnosis of thyroid cancer (4). With advances in genomic and proteomic technologies, new biomarkers for thyroid cancer have emerged. Studies have investigated the role of immunocytological markers like galectin-3 (5-7), HBME-1 (6-8), fibronectin-1, CITED-1, and cytokeratin-19 (6,7) in improving the differential diagnosis between benign and malignant thyroid nodules. However, these markers have been barely incorporated in daily routine diagnostics, mainly due to their different methodologies and considerable overlapping identification of follicular adenomas (FA) and differentiated thyroid carcinomas (DTC) (9,10). A further and important step has been achieved with the discovery of somatic mutations in about twothirds of the papillary thyroid carcinomas (PTCs) and follicular thyroid carcinomas (FTCs), offering new perspectives for the classification and diagnosis of thyroid tumors (11). Molecular testing for somatic mutations has become a promising approach and is currently the most studied molecular diagnostic method in FNAB (12,13). In most thyroid cancers, these mutations are

Figure 1. Commercially available tests and their technology over time. Arch Endocrinol Metab. 2018;62/6

577


Copyright© AE&M all rights reserved.

Molecular tests for indeterminate FNAB samples

step up from the Sanger sequencing method. Briefly, NGS can be used to sequence entire genomes or be constrained to specific areas of interest, including all 22,000 coding genes (the entire exome) or small numbers of individual genes (17). The publication of the integrated genomic characterization of PTC by The Cancer Genome Atlas (TCGA) has reduced the fraction of PTC cases with unknown oncogenic driver from 25% to 3.5% (18), offering a high potential for molecular diagnostics. The first targeted NGS panel customized for thyroid cancer was the ThyroSeq, which is commercially offered by CBLPath (Rye Brook, NY, USA) (Figure 1). In addition to the mutations detected by the 7-Gene Panel, other newly identified driver mutations in PIK3CA, TP53, TSHR, PTEN, RET, AKT1, CTNNB1, and TERT, as well as gene fusions involving BRAF, RET, NTRK1, NTRK3, AKT1, PPARG, and THADA have been added to the ThyroSeq panel (19,20). As an alternative to mutation testing, the analysis of differentially expressed genes has emerged. In this line of thought, the definition of gene expression patterns of different types of thyroid tumors has been shown to be a promising approach. The role of bioinformatics and the use of artificial models can create computer algorithms and, thus, molecular classifiers differentiating FA and FTC/follicular variant PTC to improve the differential diagnosis of cytologically indeterminate FNABs (21). Array technology has emerged as a powerful tool to assess the expression of a large number of genes. The Afirma Gene Expression Classifier (GEC) from Veracyte, Inc. (South San Francisco, CA, USA) embraces this approach by using microarray technology (Figure 1). A 167-gene classifier was developed, for which high sensitivity and negative predictive values (NPVs) have been reported (22). Using the same rationale as that of the GEC, after the first miR was discovered, miR analyses gained an important place within the study of molecular markers (23-28). The most described (at least in three studies) differentially expressed upregulated miRs in benign versus malignant nodules are miR-221, miR-222, miR146b, miR-21, miR-187, miR-197, and miR-181a (for a review on this topic, please refer to reference 29). ThyraMIR and RosettaGX Reveal are two miR panels that have been added to the market (Figure 1). The first test is performed on fresh FNAB material, while the second is performed on slide material. 578

Five tests are currently commercially available for thyroid FNABs: the new Afirma Genomic Sequencing Classifier (GSC; Veracyte, Inc., South San Francisco, CA, USA), the new version of ThyroSeq v3 (CBLPath, Inc, Rye Brook, NY, and University of Pittsburgh Medical Center, Pittsburgh, PA, USA), ThyGenX/ ThyraMIR (Interpace Diagnostics, LLC, Parsippany, NJ, USA), ThyroPrint (GeneproDX, Santiago, Chile) and Mir-THYpe (ONKOS Diagnósticos Moleculares LTDA, Ribeirão Preto, Brazil). The last two tests have not been certified by the College of American Pathologists yet, and the last is currently only available in Brazil. Recently, RosettaGX Reveal (Rosetta Genomics, Inc., Philadelphia, PA, USA) has been withdrawn from the market for undeclared reasons. However, the test became commercially available again during the period in which this review was written, and will then be described in the text.

How to evaluate the quality of a molecular test Before a test becomes commercially available, three steps are usually followed (Figure 2): the first step is to identify and define the mutation panel or the classifier using a training set. The second step is to validate the panel/classifier on a validation set, and, finally, the third and most important step is the validation of the panel/ classifier in a prospective, multicenter, and independent study. To date, most commercially available tests have been evaluated following these steps. However, the RosettaGX Reveal and the ThyGenX/ThyraMIR have not been evaluated in independent studies. Moreover, since there are no independent publications for the new AfirmaGSC and ThyroSeq v3 until this review was written, we will discuss the performance of the latest version of each test and their validation studies (Figure 2). The most comprehensively tested and evaluated commercial test is the 7-Gene Panel. After the first description of the BRAF mutation by Kimura and cols. in 2003 (14), with the recognition of the oncogenic role of the BRAF V600E mutation in approximately 58–69% of all PTCs, molecular testing for somatic mutations became an immediate approach and was the most promising molecular diagnostic tool in FNAB (15). However, genetic testing for BRAF V600E alone for the detection of PTCs is inadequate for clinical decision making, due to its low sensitivity (60%) for detecting PTCs (30). The highest sensitivity for the identification of thyroid cancers within the Arch Endocrinol Metab. 2018;62/6


Molecular tests for indeterminate FNAB samples

cytologically indeterminate category (63.7%) has been achieved by the analysis of a panel of mutations (15). The commercially available 7-Gene Panel miRInform was tested by Valderrabano and cols., who detected a mutation in 16% of 109 indeterminate nodules tested, all in Bethesda IV samples. Sensitivity and specificity in Bethesda IV specimens were 63% and 86% respectively, yielding an NPV and a positive predictive value (PPV) of 75% and 77%, respectively, and performing worse than it had done in the original study (31). In a first independent evaluation, the Afirma-GEC (32) could only identify 27% of the benign nodules instead of the 53% described by Alexander and cols. (22), and only 17% of the suspicious thyroid nodules could be confirmed to be malignant instead of the reported 38%. Therefore, the clinical applicability of the classifier showed to be questionable after this first validation study, as the NPV of the AfirmaGEC was lower than that reported by Alexander and cols. Most of these differences can be explained by the fact that PPVs and NPVs vary according to the prevalence of thyroid carcinoma in the indeterminate FNAB categories analyzed. The higher the prevalence of thyroid cancer in an indeterminate category, the higher the PPVs; and the lower the prevalence of thyroid cancer in an indeterminate category, the higher the NPVs. Twenty additional validation studies have shown a wide variability in sensitivity (75% to 100%), specificity (5% to 53%), PPV (13% to 100%), and NPV Arch Endocrinol Metab. 2018;62/6

(20% to 100%) (33). In addition to the prevalence of thyroid carcinoma in the analyzed indeterminate FNAB categories, the wide variation among the reported diagnostic values can also be explained by different defining characteristics of the study populations, such as the institutional prevalence of malignancy, sample size, Bethesda type and proportions of each Bethesda type included, the study definition of a “benign” tumor, and the predominance of Hürthle cell (HC) tumors (31). The performance of the ThyroSeq v2 panel has been evaluated in eight single-institution studies (33). In the original publication, which first validated the ThyroSeq v2, the test demonstrated a sensitivity of 90%, specificity of 93%, NPV of 96%, and PPV of 83%. Additional validation of the panel in eight studies has shown a wide range of values, including sensitivity of 40–100%, specificity of 56–93%, PPV of 13–90%, and NPV of 48–97% (for details, please refer to reference 33); these values are also lower than those first reported by Nikiforov and cols. (19,20). ThyGenX/ThyraMIR, RosettaGX Reveal, ThyroPrint, and Mir-THYpe have been validated, each in a single study, but not independently. ThyGenX/ ThyraMIR, a combination of the 7-Gene Panel with a miR expression classifier, achieved a sensitivity of 94%, specificity of 80%, NPV of 97%, and PPV of 68% (34). The RosettaGX Reveal has shown an NPV of 92%, PPV of 43%, sensitivity of 74%, and specificity of 74% 579

Copyright© AE&M all rights reserved.

Figure 2. Steps for the commercialization of a molecular test.


Molecular tests for indeterminate FNAB samples

(35). The ThyroPrint, a 10-gene classifier, has shown an NPV of 98%, PPV of 78%, sensitivity of 93%, and specificity of 81% (36). Mir-THYpe, a Brazilian 11 miRNA expression classifier, has shown an NPV of 96%, PPV of 76%, sensitivity of 95%, and specificity of 81% (37). Based on these data, the available tests can be classified as either a “rule-in” or “rule-out” test (Figure 3). Specifically, when the diagnostic test is intended to predict benign nodules (rule out), it will require a high NPV, while when intended to predict malignancy (rule in), it will require a high PPV. According to VargasSalas and cols. (38), in order to consider a test as having good rule-out ability, the test should have an NPV of at least 94%; this means that a residual risk of malignancy would be lower than 6% for a negative result, which should be close to a Bethesda II cytology. A minimum sensitivity of > 90% is necessary to keep the NPV above 94% in a broad range of disease prevalences. There is no consensus on the minimum required PPV to consider a rule-in test to be good; however, a specificity rate above 80% would result in a PPV above 60% for a disease with a prevalence rate above 25%.

Are the current tests helpful enough in diagnosing indeterminate thyroid FNAB samples?

Copyright© AE&M all rights reserved.

If we consider the NPV, PPV, sensitivity, and specificity rates mentioned before, the currently available

molecular tests would be considered as either rule-in or rule-out tests, as shown in Figure 3. In detail, the Afirma-GEC and RosettaGX Reveal could be classified as rule-out tests (based on original publication data), ThyGenX alone as a rule-in test, and ThyroSeq v2 and ThyGenX + ThyraMIR as both, rule-in and rule-out tests. Knowing the pretest malignancy risk at each test location is important to estimate the actual NPV, as discussed above; this is true particularly for rule-out tests. The pretest malignancy risk is institution-dependent and requires knowledge of the risk of malignancy for each Bethesda category (38,39). However, in daily routine, patients often bring FNAB results from various laboratories; thus, it is almost impossible to know the pretest malignancy risk for every single lab, making it hard to judge a benign test outcome of a rule-out test since the real NPV is unknown. This problem can lead to an incorrect test interpretation and wrong surgical referral. Therefore, before a molecular test is recommended to a patient, the pretest malignancy risk should be determined, and the assessment should also include other features to refine the risk level, such as ultrasound features (hypoechoic solid nodules, microcalcifications, and irregular borders) (40), cytological features (nuclear atypia, which increases the risk of malignancy) (41), and the patient’s history of radiation exposure, preferences, and family history of thyroid cancer.

Figure 3. Commercially available rule-out and rule-in tests. Afirma-GEC and RosettaGX Reveal could be classified as rule-out tests (based on original publication data), ThyGenX alone, as a rule-in test, and ThyroSeq v3 and ThyGenX + ThyraMIR as both, rule-in and rule-out tests. 580

Arch Endocrinol Metab. 2018;62/6


Further limitations of the commercially available tests must be considered. For the rule-out tests: (a) none of the validation studies (33) could confirm the initial sensitivity and specificity of the test; (b) benign samples are often not resected, thus, the number of falsenegative test outcomes are most likely underestimated and, for clinical practice, are currently unknown (42); and (c) HC tumors remain one of the main causes of incorrect tests. On the other hand, rule-in tests, in my opinion, can be somewhat a double-edged sword, since the correlation between the presence of mutations and malignancy is still imprecise in some cases. Some mutations, like BRAF V600E and TERT, are highly specific and have been well studied, showing almost a 100% risk of PTC (43,44). However, the impact of preoperatively detecting RAS mutations or PAX8/PPARg fusions is still evolving. According to Nishino and cols., the PPVs of RAS and PAX8/ PPARg for positive test outcomes among cytologically indeterminate aspirates can vary from 57% to 100% (42). Similarly, in a systematic review by Sahli and cols. including 8,162 patients, RAS mutations and RET/ PTC and PAX8/PPARg rearrangements were detected in up to 48%, 68%, and 55% of all benign nodules, respectively. Moreover, some malignant lesions showed no mutations at all (33). Since RAS mutations can be present in 30–45% of the FTCs, 30–45% of the follicular variant PTCs, 20–40% of the poorly DTCs, 10–20% of the anaplastic thyroid cancers, and 20– 25% of the FAs, their identification in molecular tests can be more confusing than helpful (45). Xing, in an excellent review, showed us the light at the end of the tunnel (45). According to his findings, RAS mutations used alone have low diagnostic sensitivity and specificity, since histologically benign nodules can be conservatively managed for a long term. However, even when histologically confirmed to be malignant, tumors positive for a RAS mutation have limited aggressiveness. Thus, based on the findings of Medici and cols. (46), Xing suggests that coexistence of a RAS mutation with additional oncogenic changes should be treated differently than the finding of a RAS mutation alone in terms of prognostic significance. If only RAS mutations are found in a DTC, it is reasonable and safe to assume a good prognosis. Therefore, there are still important technical limitations, as well as limitations with regard to the knowledge about the relevance of single mutations, Arch Endocrinol Metab. 2018;62/6

since it seems that thyroid cancer is unlikely to be accounted for by the effects of a small number of genes but by a complex interaction of multiple factors (47,48). However, with improvements in the tests, these limitations should be minimized. The technology in the new Afirma test has changed: GSC is now used, instead of GEC, in order to gain specificity and avoid histologically benign samples to be classified as suspicious. The test uses RNA-based NGS to measure gene expression, sequencing of nuclear and mitochondrial RNAs, changes in genomic copy number including loss of heterozygosity, and the development of enhanced bioinformatics and machine learning strategies. The initial validation study of the Afirma test has shown a 36% increase in specificity compared with the GEC (47). Translating into practice, at least one-third of the Bethesda III and IV nodules that are histopathologically benign will receive a benign result using the GSC compared with the GEC. The expectation is that with the 68.3% gain in specificity with the GSC, testing of Bethesda III and IV and HC neoplasms may safely reduce unnecessary surgeries. New information has also been added to ThyroSeq v3 in order to improve sensitivity. The existing test has been expanded with the incorporation of recently discovered molecular markers (new driver mutations and gene fusions), copy number variations and, also, improvement of sequencing assays, allowing the detection of multiple and various types of genetic alterations with a limited amount of cells (49). Moreover, the test’s accuracy for detecting various types of HC tumors has improved. The first results are very promising: Nikiforova and cols. demonstrated a sensitivity of 94% in the training set and 98% in the validation set (Figure 2); however, an independent, prospective, multicenter study is still needed. Despite the known limitations described here, these tests seem to be moving toward an important improvement. Still, independent validation studies of the new tests are urgently needed. Moreover, we are still waiting for solutions for other problems: high costs and challenges to send samples for analysis, especially in countries outside the Europe-US axis. In Brazil, the cost of a molecular test is almost as high as the cost of a total thyroidectomy. In addition to pricey molecular tests, import and export taxes, shipment costs, and daily variations in the dollar exchange rate have to be considered. Moreover, an excessive bureaucracy restricts the offer of the tests to one laboratory in Brazil. Patients 581

Copyright© AE&M all rights reserved.

Molecular tests for indeterminate FNAB samples


Molecular tests for indeterminate FNAB samples

interested in molecular testing have to travel to this lab to collect FNAB material. Moreover, none of the health insurances cover the costs of the tests. Thus, the application of commercial molecular tests to improve the diagnosis of cytologically indeterminate thyroid FNABs is still far from our daily clinical practice.

The Brazilian experience with molecular tests Although diagnostic parameters of sensitivity, specificity, PPV, and NPV have ​​ been published for the different available tests, each institution is required to determine its own risks of malignancy for the different indeterminate categories, particularly in regard to PPV and NPV. Only based on these local malignancy rates can a test be defined as being good or not in terms of local PPVs and NPVs. Based on that, I conducted a survey of all tests performed in Brazil using commercially available assays to evaluate their usefulness and performance as rule-in and rule-out tests in the country. The results of the survey are detailed below. Molecular tests have been available in Brazil since 2013. The first commercialized test was miRInform, and a total of seven tests were performed in the country before the test was replaced with ThyGenX. The ThyGenX/ ThyraMIR was commercialized for a short period of time, and only three ThyGenX tests were performed. Currently, only the ThyroSeq v2 (and now the ThyroSeq v3) test is commercialized in Brazil (by the laboratory Cytolog), since the RosettaGX Reveal has been recently removed from the market. Overall, 13

ThyroSeq v2, 4 ThyroSeq v3, and 5 RosettaGX Reveal tests have been performed (Table 1). The Afirma test was previously commercialized by the laboratory Fleury but is no longer available. Because thyroid nodules with mutation-negative (ThyroSeq v2 and v3) or benign results when tested with Afirma-GEC and RosettaGX Reveal are rarely resected, the false-negative rates and the sensitivity and specificity values of these tests in our statistics are currently unknown. Of all, 8 out of 17 samples analyzed with either the ThyroSeq v2 or the ThyroSeq v3 showed a positive result (presence of mutation); on histology, 4 of these samples with a positive result confirmed to be malignant on histology, while 2 were benign (1 was an FA with a THADA/ IGF2BP3 fusion, and 1 was negative for malignancy and had associated Hashimoto’s thyroiditis with the presence of MET gene overexpression) and 2 have still not been operated on. Of the 5 samples evaluated with the RosettaGX Reveal, 3 were suspect for malignancy (1 was confirmed be malignant on histology), while 2 have not been operated yet. Unfortunately, the Afirma-GEC data for the Brazilian population was not accessible due to the ethical policy of the Fleury laboratory. Due to limited follow-up data, the sensitivity, specificity, and accuracy of these tests could not be determined for the Brazilian population and still remain to be evaluated. Moreover, as suggested by different groups, it is strictly important for each local setting to assess their own pretest probability of malignancy.

Table 1. Description of all molecular tests commercially available in Brazil and performed to the current date to evaluate nodules deemed cytologically indeterminate on fine-needle aspiration biopsy

Copyright© AE&M all rights reserved.

Test

Result

Bethesda

Histology

ThyroSeq v2

Positive (THADA/IGF2BP3 fusion)

SFN (Bethesda IV)

Follicular adenoma

ThyroSeq v2

Positive (NRAS + TERT)

AUS (Bethesda III)

Papillary carcinoma

ThyroSeq v2

Positive (multiple gene expression with abnormalities)

SFN + HC (Bethesda IV)

Papillary carcinoma, oncocytic variant

ThyroSeq v2

Positive (BRAF K601)

AUS

Papillary carcinoma, follicular variant

ThyroSeq v2

Positive (overexpression of the MET gene)

AUS

Negative for malignancy, Hashimoto’s thyroiditis

ThyroSeq v2

Positive (overexpression of the MET gene)

AUS

NO

ThyroSeq v2

7 negative

4 AUS / 3 SFN

NO

ThyroSeq v3

Positive (KRAS + copy number + gene expression alteration)

SFN + HC (Bethesda IV)

Multifocal micropapillary carcinoma

ThyroSeq v3

Positive (copy number alterations)

SFN + HC (Bethesda IV)

NO

ThyroSeq v3

2 negative

1 AUS / 1 SFN

NO

RosettaGX Reveal

Suspect

SFN

Papillary carcinoma, solid variant

RosettaGX Reveal

2 suspect

2 SFN

NO

RosettaGX Reveal

2 benign

1 AUS / 1 SFN + HC

NO

SFN: suspicious for follicular neoplasm; AUS: atypia of undetermined significance; HC: Hürthle cell tumor; NO: not operated.

582

Arch Endocrinol Metab. 2018;62/6


Molecular tests for indeterminate FNAB samples

As mentioned earlier, although these molecular tests are available in Brazil, due to their high costs, they still have a limited applicability in daily clinical practice. The Mir-THYpe test was developed and validated by the startup ONKOS Diagnósticos Moleculares LTDA in partnership with the Cancer Hospital of Barretos. This is the only genetic test for molecular classification of indeterminate thyroid nodules that has been developed and validated exclusively in Brazilian patients. The MirTHYpe analyzes genetic material (miRs) extracted from previously collected FNAB slides. The results of the Mir-THYpe validation study showed an NPV of 96%, PPV of 76%, sensitivity of 94.6%, and specificity of 81% (37), within a range of pretest probability of malignancy of 39%. With these numbers, the test showed to have both optimal rule-in and rule-out performances and to be safe in all three indeterminate categories: III (AUS/ FLUS), IV (suspicious for follicular neoplasm), and V (suspicious for malignancy). These are promising data, especially for our population. However, a multicenter, independent study and longitudinal follow-up studies of unresected nodules with negative molecular testing results are strongly recommended. In conclusion, while some tests have been withdrawn from the market, it seems that Afirma-GSC and ThyroSeq v3 are well-established tests that can be used as another tool in addition to clinical, radiological, and cytological features to help diagnose indeterminate FNAB samples. The initial sensitivity and specificity rates of the previous versions of the tests lack confirmation, and the tests underestimate false-negative cases; additionally, HC tumors remain one of the major causes of incorrect tests. Moreover, some somatic mutations still need more clarification in regard to their functional characterization and impact on tumor aggressiveness. The new versions of both tests, Afirma-GSC and ThyroSeq v3, show improved sensitivity and specificity but still require further independent validation. Still, local malignancy rates need to be determined, and more independent validation studies must be performed, in addition to studies comparing the performance of each test, although this may be difficult due to the individual characteristics and objectives of each test. Finally, we are still waiting for validation results of new tests (ThyroPrint and Mir-THYpe), since these tests have a much lower cost and, therefore, will be more accessible to patients in Latin America and Brazil.

ding me with the data of the molecular tests and making every effort to obtain the follow-up data of the patients. I am also very thankful to Prof. Dr. Markus Eszlinger for his valuable comments in this review article.

Acknowledgments: I am grateful to Dr. Fabiano Callegary, pathologist and owner of the Cytolog Lab, for generously provi-

16. Nikiforov YE, Steward DL, Robinson-Smith TM, Haugen BR, Klopper JP, Zhu Z, et al. Molecular testing for mutations in improving

Arch Endocrinol Metab. 2018;62/6

Disclosure: no potential conflict of interest relevant to this article was reported.

1. Carpi A, Ferrari E, Toni MG, Sagripanti A, Nicolini A, Di Coscio G. Needle aspiration techniques in preoperative selection of patients with thyroid nodules: a long-term study. J Clin Oncol. 1996;14:1704–12. 2. Cibas ES, Ali SZ. NCI Thyroid FNA State of the Science Conference. The Bethesda System for Reporting Thyroid Cytopathology. Am J Clin Pathol. 2009;132(5):658-65. 3. Cibas ES, Ali SZ. The 2017 Bethesda System for Reporting Thyroid Cytopathology. Thyroid. 2017;27(11):1341-6. 4. Melvin KE, Miller HH, Tashjian AH Jr. Early diagnosis of medullary carcinoma of the thyroid gland by means of calcitonin assay. N Engl J Med. 1971;285(20):1115-20. 5. Bartolazzi A, Orlandi F, Saggiorato E, Volante M, Arecco F, Rossetto R, et al. Galectin-3-expression analysis in the surgical selection of follicular thyroid nodules with indeterminate fine-needle aspiration cytology: a prospective multicentre study. Lancet Oncol. 2008;9:543-49. 6. de Matos PS, Ferreira AP, de Oliveira Facuri F, Assumpcao LV, Metze K, Ward LS. Usefulness of HBME-1, cytokeratin 19 andgalectin-3 immunostaining in the diagnosis of thyroid malignancy. Histopathology. 2005;47:391-401. 7. Saggiorato E, De Pompa R, Volante M, Cappia S, Arecco F, Dei Tos AP, et al. Characterization of thyroid ‘follicular neoplasms’ in fine-needle aspiration cytological specimens using a panel of immunohistochemical markers: a proposal for clinical application. Endocr Relat Cancer. 2005;12:305-17. 8. Volante M, Bozzalla-Cassione F, DePompa R, Saggiorato E, Bartolazzi A, Orlandi F, et al. Galectin-3 and HBME-1 expression in oncocytic cell tumors of the thyroid. Virchows Arch. 2004;445:183-8. 9. Faggiano A, Caillou B, Lacroix L, Talbot M, Filetti S, Bidart JM, et al. Functional characterization of human thyroid tissue with immunohistochemistry. Thyroid. 2007;17:203-11. 10. Freitas BC, Cerutti JM. Genetic markers differentiating follicular thyroid carcinoma from benign lesions. Mol Cell Endocrinol. 2010;321:77-85. 11. Eszlinger M, Krohn K, Hauptmann S, Dralle H, Giordano TJ, Paschke R. Perspectives for improved and more accurate classification of thyroid epithelial tumors. J Clin Endocrinol Metab. 2008;93:3286-94. 12. Moses W, Weng J, Sansano I, Peng M, Khanafshar E, Ljung BM, et al. Molecular testing for somatic mutations improves the accuracy of thyroid fine-needle aspiration biopsy. World J Surg. 2010;34:2589-94. 13. Eszlinger M, Paschke R. Molecular fine-needle aspiration biopsy diagnosis of thyroid nodules by tumor specific mutations and gene expression patterns. Mol Cell Endocrinol. 2010;322:29-37. 14. Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 2003;63(7):1454-7. 15. Ferraz C, Eszlinger M, Paschke R. Current state and future perspective of molecular diagnosis of fine-needle aspiration biopsy of thyroid nodules. J Clin Endocrinol Metab. 2011;96:2016-26.

583

Copyright© AE&M all rights reserved.

REFERENCES


Molecular tests for indeterminate FNAB samples

the fine-needle aspiration diagnosis of thyroid nodules. J Clin Endocrinol Metab. 2009;94:2092-8. 17. Behjati S, Tarpey PS. What is next generation sequencing? Arch Dis Child Educ Pract Ed. 2013;98:236-38. 18. Network CGAR. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159(3):676-90. 19. Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, et al. Highly accurate diagnosis of cancer in thyroid nodules with follicular neoplasm/suspicious for a follicular neoplasm cytology by ThyroSeq v2 next-generation sequencing assay. Cancer. 2014;120(23):3627-34. 20. Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, et al. Impact of the Multi-Gene ThyroSeq Next-Generation Sequencing Assay on Cancer Diagnosis in Thyroid Nodules with Atypia of Undetermined Significance/Follicular Lesion of Undetermined Significance Cytology. Thyroid. 2015;25(11):1217-23. 21. Libbrecht MW, Noble WS. Machine learning in genetics and genomics. Nat Rev Genet. 2015;16(6):321-32. 22. Alexander EK, Kennedy GC, Baloch ZW, Cibas ES, Chudova D, Diggans J, et al. Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N Engl J Med. 2012;367(8):705-15. 23. Pallante P, Visone R, Ferracin M, Ferraro A, Berlingieri MT, Troncone G, et al. MicroRNA deregulation in human thyroid papillary carcinomas. Endocr Relat Cancer. 2006;13:497-508. 24. He H, Jazdzewski K, Li W, Liyanarachchi S, Nagy R, Volinia S, et al. The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci USA. 2005;102:19075-80. 25. Mazeh H, Mizrahi I, Halle D, Ilyayev N, Stojadinovic A, Trink B, et al. Development of a microRNA-based molecular assay for the detection of papillary thyroid carcinoma in aspiration biopsy samples. Thyroid. 2011;21:111-8. 26. Tetzlaff MT, Liu A, Xu X, Master SR, Baldwin DA, Tobias JW, et al. Differential expression of miRNAs in papillary thyroid carcinoma compared to multinodular goiter using formalin fixed paraffin embedded tissues. Endocr Pathol. 2007;18:163-73. 27. Nikiforova MN, Chiosea SI, Nikiforov YE. MicroRNA expression profiles in thyroid tumors. Endocr Pathol. 2009;20:85-91. 28. Vriens MR, Weng J, Suh I, Huynh N, Guerrero MA, Shen WT, et al. MicroRNA expression profiling is a potential diagnostic tool for thyroid cancer. Cancer 2012;118:3426-32. 29. Lodewijk L, Prins AM, Kist JW, Valk GD, Kranenburg O, Rinkes IH, et al. The value of miRNA in diagnosing thyroid cancer: a systematic review. Cancer Biomark. 2012;11:229-38. 30. Pusztaszeri MP, Krane JF, Faquin WC. BRAF testing and Thyroid FNA. Cancer Cytopathol. 2015;123(12):689-95. 31. Valderrabano P, Leon ME, Centeno BA, Otto KJ, Khazai L, McCaffrey JC, et al. Institutional prevalence of malignancy of indeterminate thyroid cytology is necessary but insufficient to accurately interpret molecular marker tests. Eur J Endocrinol. 2016;174:621-9. 32. McIver B, Castro MR, Morris JC, Bernet V, Smallridge R, Henry M, et al. An Independent Study of a Gene Expression Classifier (Afirma) in the Evaluation of Cytologically Indeterminate Thyroid Nodules. J Clin Endocrinol Metab. 2014;99:4069-77.

Copyright© AE&M all rights reserved.

33. Sahli ZT, Smith PW, Umbricht CB, Zeiger MA. Umbricht and Martha A. Zeiger. Preoperative Molecular Markers in Thyroid Nodules. Front Endocrinol (Lausanne). 2018;9:179. 34. Labourier E, Shifrin A, Busseniers AE, Lupo MA, Manganelli ML, Andruss B, et al. Molecular Testing for miRNA, mRNA, and DNA

584

on Fine-Needle Aspiration Improves the Preoperative Diagnosis of Thyroid Nodules With Indeterminate Cytology. J Clin Endocrinol Metab. 2015;100(7):2743-50. 35. Lithwick-Yanai G, Dromi N, Shtabsky A, Morgenstern S, Strenov Y, Feinmesser M, et al. Multicentre validation of a microRNA-based assay for diagnosing indeterminate thyroid nodules utilising fine needle aspirate smears. J Clin Pathol. 2017;70:500-7. 36. Gonzalez HE, Martınez JE, Vargas-Salas S, Solar A, Veliz L, Cruz F, et al. A 10-Gene Classifier for Indeterminate Thyroid Nodules: Development and Multicenter Accuracy Study. Thyroid. 2017;27(8):1058-67. 37. dos Santos MT, Buzolin AL, Gama RB, da Silva ECA MD, Dufloth RM, Figueiredo DLA, et al. Molecular classification of thyroid nodules with indeterminate cytology: development and validation of a highly sensitive and specific new miRNA-based classifier test using fine-needle aspiration smear slides. Thyroid. 2018: Oct 15. doi: 10.1089/thy.2018.0254. [Epub ahead of print] 38. Vargas-Salas S, Martínez JR, Urra S, Domínguez JM, Mena N, Uslar T, et al. Genetic testing for indeterminate thyroid cytology: review and meta-analysis. Endocr Relat Cancer. 2018;25(3):R163-77. 39. Valderrabano P, McIver B. Evaluation and Management of Indeterminate Thyroid Nodules: The Revolution of Risk Stratification Beyond Cytological Diagnosis. Cancer Control. 2017;24(5):1-14. 40. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016;26(1):1-133. 41. Walts AE, Mirocha J, Bose S. Follicular lesion of undetermined significance in thyroid FNA revisited. Diagn Cytopathol. 2014;42(1):18-22. 42. Nishino M. Molecular cytopathology for thyroid nodules: A review of methodology and test performance. Cancer Cytopathol. 2016;124(1):14-27. 43. Liu R, Xing M. Diagnostic and prognostic TERT promoter mutations in thyroid fine-needle aspiration biopsy. Endocr Relat Cancer. 2014;21:825-30. 44. Xing M, Liu R, Liu X, Murugan AK, Zhu G, Zeiger MA, et al. BRAF V600E and TERT promoter mutations cooperatively identify the most aggressive papillary thyroid cancer with highest recurrence. J Clin Oncol. 2014;32:2718-26. 45. Xing M. Clinical utility of RAS mutations in thyroid cancer: a blurred picture now emerging clearer. BMC Medicine. 2016;14:12. 46. Medici M, Kwong N, Angell TE, Marqusee E, Kim MI, Frates MC, et al. The variable phenotype and low-risk nature of RAS-positive thyroid nodules. BMC Med. 2015;13:184. 47. Patel KN, Angell TE, Babiarz J, Barth NM, Blevins T, Duh QY, et al. Performance of a Genomic Sequencing Classifier for the Preoperative Diagnosis of Cytologically Indeterminate Thyroid Nodules. JAMA Surg. 2018;153(9):817-24. 48. Herceg Z, Hainaut P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Mol Oncol. 2007;1(1):26-41. 49. Nikiforova MN, Mercurio S, Wald AI, Barbi de Moura M, Callenberg K, Santana-Santos L, et al. Analytical performance of the ThyroSeq v3 genomic classifier for cancer diagnosis in thyroid nodules. Cancer. 2018;124(8):1682-90.

Arch Endocrinol Metab. 2018;62/6


original article

The characteristics of blood glucose fluctuations in patients with fulminant type 1 diabetes mellitus in the stable stage Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China 2 Division of Endocrinology, Department of Medicine, National University of Singapore, Singapore 3 National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore * Contributions were equal to those of first author 1

Jie Wang1,*, Bing-Li Liu1,*, Zheng Li1, Hui-Qin Li1, Rui Sun1, Yun Hu1, Kok-Onn Lee2, Lei Ye3, Xiao-Fei Su1, Jian-Hua Ma1

ABSTRACT Objective: The aim was to characterize blood glucose fluctuations in patients with fulminant type 1 diabetes (FT1DM) at the stable stage using continuous blood glucose monitoring systems (CGMSs). Subjects and methods: Ten patients with FT1DM and 20 patients with classic type 1 diabetes mellitus (T1DM) (the control group) were monitored using CGMSs for 72 hours. Results: The CGMS data showed that the mean blood glucose (MBG), the standard deviation of the blood glucose (SDBG), the mean amplitude glycemic excursions (MAGE), the blood glucose areas and the percentages of blood glucose levels below 13.9 mmol/L were similar between the two groups. However, the percentage of blood glucose levels below 3.9 mmol/L was significantly higher in the FT1DM group compared to the T1DM group (p < 0.05). The minimum (Min) blood glucose level in the FT1DM group was significantly lower than that of the T1DM group (p < 0.05). Patients with FT1DM had severe dysfunction of the islet beta cells and alpha cells compared to patients with T1DM, as indicated by lower C-peptide values and higher glucagon/C-peptide values. Conclusion: In conclusion, patients with FT1DM at the stable stage were more prone to hypoglycemic episodes as recorded by CGMSs, and they had a greater association with severe dysfunction of both the beta and alpha islet cells compared to patients with T1DM. Arch Endocrinol Metab. 2018;62(6):585-90 Keywords Fulminant type 1 diabetes; type 1 diabetes; continuous glucose monitoring system (CGMS)

T

ype 1 diabetes mellitus (T1DM) is characterized by insulin deficiency and the destruction of pancreatic beta cells. It is mainly classified into classic T1DM and fulminant type 1 diabetes mellitus (FT1DM). FT1DM is a special subtype of type 1 diabetes first proposed by Imagawa and cols. in 2000 (1). FT1DM is characterized by an abrupt onset, dangerous conditions, ketosis or ketoacidosis at the time of diagnosis, high blood glucose levels, lower glycosylated hemoglobin (HbA1c) levels, serious destruction of pancreatic beta cells, and negative islet-related autoantibodies, such as insulin antibodies, anti-GAD antibodies (GADAb), and insulin autoantibodies (IAA). In addition, most patients with FT1DM report having had symptoms of influenza or gastrointestinal symptoms prior to onset (2). Recently, reports on FT1DM have increased, especially in East Asia (2-6). However, detailed characteristics of glucose fluctuations in patients with FT1DM at the stable stage are unclear. Arch Endocrinol Metab. 2018;62/6

Jian-Hua Ma Department of Endocrinology Nanjing Medical University Affiliated Nanjing Hospital 210006 – Nanjing China majianhua@china.com Xiao-Fei Su Department of Endocrinology Nanjing Medical University Affiliated Nanjing Hospital 210006 – Nanjing China suxiaofeifei@126.com Received on Jan/12/2018 Accepted on Jun/14/2018 DOI: 10.20945/2359-3997000000082

The current study aimed to utilize CGMSs to characterize blood glucose fluctuations in patients with FT1DM at the stable stage.

SUBJECTS AND METHODS Subjects This was a retrospective study. We identified 10 patients with FT1DM at the stable stage and 20 patients with classic T1DM as control patients from Jan. 2012 to Dec. 2015 at Nanjing First Hospital, Nanjing, Jiangsu, China. The FT1DM group included 6 female and 4 male patients, and their mean history of T1DM was 7.20 ± 5.83 years. All of the patients with FT1DM met the diagnostic criteria proposed by Imagawa: 1) ketosis or ketoacidosis that occurred in a high glucose state, usually less than 1 week; 2) plasma glucose levels > 16 mmol/L (> 288 mg/dL) and HbA1c levels < 8.7% in newly diagnosed patients; 3) excretion of urinary 585

Copyright© AE&M all rights reserved.

INTRODUCTION

Correspondence to:


Blood glucose fluctuations in T1DM

C-peptide < 10 ug/dL with a fasting plasma C-peptide (C-P) < 0.1 nmol/L (0.3 ug/L) and stimulation plasma C-peptide (after a meal or glucagon injection) < 0.17 nmol/L (0.5 µg/L) (7). The T1DM group included 8 female and 12 male patients, and their mean history of T1DM was 9.60 ± 8.40 years. The diagnosis of T1DM was based on the criteria for classical T1DM introduced by the WHO in 1999 (8). The protocol and informed consent documents were approved by the Institutional Ethics Committee at Nanjing First Hospital. All of the patients gave written informed consent.

Copyright© AE&M all rights reserved.

Methods Demographic information about the patients was collected, including each patient’s age, history of diabetes, BMI, waist circumference, hip circumference, waist-hip ratio, and insulin dose. Continuous glucose data were obtained using the Medtronic Minimed CGMS Gold (Medtronic Incorporated, Northridge, USA) for at least 3 days after being admitted to the hospital for 2 days. The CGMS induction probe detected the concentration of abdominal subcutaneous interstitial fluid glucose in the subjects. It was connected to a recorder via a cable so that the glucose concentrations could be obtained and recorded every 5 minutes (min). A total of 288 glucose readings were recorded automatically every day for three days, and the range of blood glucose levels recorded was 2.2-22.2 mmol/L (39.6-399.6 mg/dL). Peripheral blood glucose levels were measured at least 4 times per day during the monitoring period to calibrate the CGMS results. The data obtained from the CGMSs included the following parameters: the 24-hour (hr) mean blood glucose level (MBG), the standard deviation of the MBG level (SDBG), the 24-hr mean amplitude of glycemic excursions (MAGE), the percentage time durations (%) of hyperglycemia (glucose > 13.3 mmol/L) (> 239.4 mg/dL) and hypoglycemia (glucose < 3.9 mmol/L) (< 70.2 mg/dL), the maximum blood glucose level (Max) and the minimum blood glucose level (Min). The clinical and biochemical parameters included the following: fasting C-peptide (C-P0), 2-hr postprandial C-peptide (C-P2hr), fasting glucagon (Glu0), 2-hr postprandial glucagon (Glu2hr), HbA1c, anti-GAD or insulin antibodies, blood routine, biochemical indexes, and calculations for fasting glucagon/ fasting C-peptide (Glu0/C-P0) and 2-hr postprandial 586

glucagon/2-hr postprandial C-peptide (Glu2hr/CP2hr). Plasma glucose levels were measured using the glucose oxidase method. C-peptide was measured using a chemiluminescent immunometric assay on the Modular Analytics E170 (Roche® Diagnostics GmbH, Mannheim, Germany). Glucagon levels were determined using a quantitative radioimmunoprecipitation assay kit (Beijing North Institute of Biological Technology, China). HbA1c levels were measured using a highperformance liquid chromatography (HPLC) assay (Bio-Rad Laboratories, Inc. CA, USA). The breakfast for the patients’ OGTT was 250 ml milk, 0.1 kg steamed bread meal, and an egg.

Statistical analysis For normally distributed data, the means ± standard deviations (SDs) of the two groups were compared using t-tests. For data that were nonnormally distributed, the medians and interquartile ranges were compared using nonparametric Mann-Whitney U tests. The rates between the two groups were compared using the chisquared test. Significance was defined as p < 0.05.

RESULTS Patient demographics There were no significant differences in the genders and histories of diabetes between the two groups. Ages, BMIs, and waist-to-hip ratios in the FT1DM group were higher than those in the T1DM group (p < 0.05 for all); premeal insulin doses (unit/kg/ day) were significantly higher in the FT1DM group compared to the T1DM group (p < 0.05). Daily doses of insulin (unit/kg/day) and basal doses of insulin (unit/kg/day) were similar between the two groups (p > 0.05) (Table 1).

Laboratory measurements Patients with FT1DM had lower C-P0 and C-P2hr levels and higher Glu2hr, Glu0/C-P0, and Glu2hr/C-P2hr levels than those with T1DM (p < 0.05), suggesting that the regulation of glucagon secretion from α cells is impaired in FT1DM. GAD antibody levels in the FT1DM group were significantly lower than in the T1DM group (p < 0.05). There were no significant differences in HbA1c, INS-Ab, and Glu0 levels between the two groups (p > 0.05 for all) (Table 1). Arch Endocrinol Metab. 2018;62/6


Blood glucose fluctuations in T1DM

Blood urea nitrogen (BUN) and alkaline phosphatase levels were significantly higher in the FT1DM group than in the T1DM group (p < 0.05). There were no significant differences in the blood routines (white blood cells, red blood cells, hemoglobin, and platelet count) or biochemical

indexes (alanine aminotransferase, albumin, globulin, potassium, sodium, chlorine, cholesterol, triglyceride, high density lipoprotein, glutamine transpeptidase, blood creatinine, urinary albumin, and uric acid) between the two groups (p > 0.05 for all) (Table 1).

Table 1. Clinical and biochemical characteristics FT1DM (n = 10)

T1DM (n = 20)

t/z/χ²

P

Gender (%)

4/6

12/8

1.071

0.442

Age (years)

43 (37.5, 54)

35 (26.75, 38.75)

-2.312

0.021*

History (years)

6 (3.25, 10.5)

9.5 (2.25,17)

-0.596

0.551

Body mass index (kg/m2)

23.01 ± 2.64

20.98 ± 2.01

2.348

0.026*

Wasit to hip ratio (%)

0.98 ± 0.04

0.93 ± 0.06

2.454

0.021*

The basal dosages of insulin (u/kg/day)

0.24 ± 0.08

0.27 ± 0.15

-0.582

0.565

0.31 (0.26, 0.61)

0.24 (0.17, 0.31)

-2.268

0.023*

0.63 ± 0.20

0.52 ± 0.18

1.657

0.109

Fasting C-peptide (µg/L)

0.01 (0.01, 0.04)

0.07 (0.01, 0.20)

-2.227

0.026*

2-hr postprandial C-peptide (µg/L)

0.01 (0.01, 0.04)

0.11 (0.02, 0.52)

-2.778

0.005**

Fasting glucagon (ng/L)

156.06 ± 43.86

136.19 ± 28.03

1.512

0.142

The pre-meal dosages of insulin (u/kg/day) The daily dosages of Insulin (u/kg/day)

208.74 (166.39, 271.73)

165.31 (144.93, 200.39)

-2.202

0.028*

Fasting glucagon/Fasting C-peptide (%)

13271.00 (5193.94, 18294.25)

2227.86 (627.63, 13705.75)

-2.024

0.043*

2-hr postprandial glucagon/2-hr postprandial C-peptide (%)

17595.50 (9020.94, 26907.00)

1339.37 (291.14, 7466.25)

-3.08

0.002**

8.03 ± 1.31

9.70 ± 2.65

-1.864

0.073*

Anti-GAD antibodies (%)

5.48 (4.67, 7.21)

13.40 (9.36, 23.03)

-3.696

0.000**

Anti-Insulin antibodies (%)

3.37 (2.86, 8.26)

6.09 (2.95, 11.62)

-1.16

0.246

White blood cells (*10 /L)

6.02 ± 3.51

6.88 ± 2.83

-0.728

0.473

4.12 ± 0.44

4.42 ± 0.49

-1.632

0.114

156.8 ± 56.13

196.05 ± 63.82

-1.649

0.110

2-hr postprandial glucagon (ng/L)

Glycosylated hemoglobin (%)

9

Red blood cells (*10 /L) 12

Platelet count (*109/L) Haemoglobin (g/L)

131 ± 9.24

134.85 ± 18.51

-0.617

0.543

25.00 (16.50, 38.75)

19.00 (11.00, 24.75)

-1.652

0.099

Albumin (g/L)

41.86 ± 9.67

42.63 ± 3.59

-0.317

0.754

Globulin (g/L)

25.32 ± 3.76

25.49 ± 4.77

-0.098

0.922

Alanine aminotransferase (U/L)

Potassium (mmol/L)

4.05 (3.71, 4.27)

4.42 (3.79, 4.74)

-1.651

0.099

Sodium (mmol/L)

141.05 ± 4.87

138.94 ± 4

1.269

0.215

Chloride (mmol/L)

102.97 ± 5.59

102.15 ± 4.08

0.461

0.648

Cholesterol (mmol/L)

4.66 ± 0.77

5.26 ± 0.94

-1.736

0.094

Triglyceride (mmol/L)

1.12 ± 0.78

1.03 ± 0.71

0.315

0.755

HDL-cholesterol (mmol/L) Glutamyl transpeptidase (U/L) Alkaline phosphatase (U/L) Urinary albumin (%) Blood urea nitrogen (mmol/L) Uric Acid (umol/L) Blood creatinine (mmol/L)

1.5 ± 0.51

1.77 ± 0.53

-1.344

0.190

18.5 (12.00, 27.75)

15.50 (12.00, 24.50)

-0.794

0.427

98.70 ± 25.27

72.25 ± 18.00

3.312

0.003**

3/7

2/18

1.92

0.300

6.66 (6.18, 10.65)

5.50 (3.97, 6.55)

-2.376

0.017*

375.50 (141.75, 519.25)

304 (253, 391)

-0.311

0.755

86.00 (62.75, 117.50)

68.7 (54.08, 74.95)

-1.716

0.086

Copyright© AE&M all rights reserved.

Groups

Normally distributed data was compared using t-test. Non-normally distributed data was compared using the nonparametric Mann-Whitney test. The rates between the two groups were compared using the chi-square test. * = p < 0.05 , ** = p < 0.01. Arch Endocrinol Metab. 2018;62/6

587


Blood glucose fluctuations in T1DM

CGMS results The CGMS data showed that the percentage of blood glucose levels < 3.9 mmol/L was higher in the FT1DM group than in the T1DM group (p < 0.05). The Min blood glucose level in the FT1DM group was lower than the Min in the T1DM group (p < 0.05) (Table 2). Other CGMS parameters (MBG, SDBG, MAGE, Max and the percentage of blood glucose levels > 13.9 mmol/L) were similar between the two groups (p > 0.05 for all).

DISCUSSION In the present study, CGMS data showed that patients in the FT1DM group had a higher risk of hypoglycemia at the stable stage, worse destruction of pancreatic beta cells, and more severe dysfunction of the islet alpha cells than patients with T1DM. Renal dysfunction was more commonly found in patients with FT1DM. Other CGMS parameters did not differ significantly between the two groups. There were no significant differences found in MBG, SDBG, MAGE and Max levels between the FT1DM and T1DM patients. These results indicate that blood glucose fluctuations were similar in the FT1DM and T1DM groups. However, the percentage of blood glucose levels < 3.9 mmol/L was higher and the Min blood glucose level was lower in the FT1DM group compared to the T1DM group (p < 0.05 for both). These findings suggest that patients with FT1DM have a higher risk of hypoglycemia compared to patients with T1DM. A 5-year follow-up study of 41 patients

with FT1DM performed by Murase and cols. showed that blood glucose fluctuations (measured using 7-point blood glucose monitoring) and the frequencies of severe hypoglycemia were higher in a group of FT1DM patients than in a group of T1DM patients (9). The results of the current study partially agree with this, as we only found that patients with FT1DM had a higher risk of hypoglycemia compared to patients with T1DM, which was indicated by the higher percentage of blood glucose levels < 3.9 mmol/L and the lower minimum blood glucose levels in the FT1DM group. However, we did not find significant differences among other indexes for blood glucose fluctuations between the two groups. This difference may be associated with the methods that were used to monitor blood glucose levels. We used CGMSs to monitor blood glucose levels, which are more accurate and occur in real-time (10). We also noticed that our patients with FT1DM were older and had higher BMIs and waist-to-hip ratios than the T1DM patients. The finding of higher BMIs in patients with FT1DM compared with those with T1DM was consistent with results of previous studies (2,11-13). The higher BMIs in patients with FT1DM compared with those with T1DM may be due to the rapid onset and short duration of FT1DM, which does not reduce body weight significantly. However, beta cell function was severely damaged, which led to lower C-peptide levels in patients with FT1DM. Furthermore, we observed that premeal doses of insulin (unit/kg/day) were significantly higher in the FT1DM group than in the T1DM group. However, while the total daily doses of insulin were higher in the FT1DM group, there was no significant difference between the two groups. One possible reason for

Table 2. CGMS data FT1DM (n = 10)

T1DM (n = 20)

95% confidence interval

t/z

P

Mean blood glucose (mmol/L)

8.86 ± 3.21

11.39 ± 3.36

(-5.1637, 0.0917)

-1.977

0.058

Standard deviation of blood glucose (mmol/L)

3.47 ± 1.07

3.12 ± 1.25

(-0.5912, 1.3012)

0.769

0.449

Mean amplitude glycemic excursions (mmol/L)

9.56 ± 2.77

7.76 ± 3.72

(-0.9354, 4.5213)

1.346

0.189

8.00 (2.00, 49.00)

15.50 (1.50, 57.50)

/

-0.529

0.597

0.10 (0, 1.15)

0.325 (0, 2.05)

/

-0.880

0.379

16.00 (0, 34.50)

0 (0, 0)

/

-2.979

0.003**

Indexes

Percentage of time above 13.9 (PT1)

Copyright© AE&M all rights reserved.

AUC > 13.9 mmol/L Percentage of time below 3.9 (PT2) AUC < 3.9 mmol/L

0.10 (0, 0.20)

0 (0, 0)

/

-3.388

0.001**

The maximum (mmol/L)

16.16 ± 3.38

17.27 ± 4.40

(-4.3635, 2.1415)

-0.700

0.490

The minimum (mmol/L)

2.40 (2.20, 5.03)

5.15 (3.83, 6.55)

/

-2.360

0.018*

AUC = Area under curve. Normally distributed data was compared using t-test. Non- normally distributed data was compared using the nonparametric Mann-Whitney test. The rates between the two groups were compared using the chi-square test.* = p < 0.05,** = p < 0.01.

588

Arch Endocrinol Metab. 2018;62/6


the pre-meal doses of insulin may be related to the more severely damaged islet function in patients with FT1DM, i.e., the poor function of islet beta cells and the dysfunction of islet alpha cells, resulting in a need for more insulin to reduce the postprandial blood glucose level. Imagawa and cols. found that daily doses of insulin were significantly higher in 161 FT1DM patients than in T1DM patients in follow-ups at 3, 6, and 12 months (11). This is different from the results of our study, and this difference may be related to the sample size, as we only studied 10 patients whose total daily doses of insulin had a tendency to be higher (0.63 ± 0.20 u/kg/day) than those of T1DM patients (0.52 ± 0.18 u/kg/day, p = 0.109). Islet function was more severely damaged in FT1DM patients, as they had lower C-P0 and C-P2hr levels and higher Glu2hr, Glu0/C-P0, and Glu2hr/ C-P2hr levels compared with T1DM patients (p < 0.05). Though the pathogenesis of FT1DM is currently unclear, it may be mediated by a variety of factors: viral infections, pregnancy, drugs, etc., in addition to autoimmune and genetic factors that can trigger an accelerated immune reaction and promote massive beta-cell death in genetically susceptible individuals. In contrast, a small number of islet cells may still be viable in T1DM patients at the onset of the disease (2,6,14,15). Zheng and cols. proposed that the appearance of a low titer of autoantibodies was likely caused by the viral infection destroying the islet cells (3). Shibasaki and cols. suggested that FT1DM is associated with viral infections and is affected by the patient’s genetic background (16). However, Imagawa and cols. found that although GAD-AB, INS-AB, and other related antibodies appeared in patients with FT1DM, they were in low antibody titers and appeared in a short-term national survey (11). Studies have suggested that intestinal viruses and chemokines not only damage the islet beta cells but also accelerate the autoimmune reaction, which destroys the remaining islet beta cells (17,18). Our study found that the GAD-AB was significantly lower in the FT1DM group than in the T1DM group, while the INS-AB titer findings were similar between the two groups. This is consistent with previous reports (3,9). Patients with FT1DM had lower C-P0 and C-P2hr levels than patients with T1DM in our study, which could be related to the stage of onset of disease (2,6). This suggests that the function of islet beta cells in FT1DM was rapidly damaged from the onset Arch Endocrinol Metab. 2018;62/6

of the disease, progressively declined, and could still be declining in T1DM after a few years. Meanwhile, patients with FT1DM had higher Glu2hr, Glu0/C-P0, and Glu2hr/C-P2hr levels than those with T1DM, suggesting that glucagon secretion was also damaged in the FT1DM group compared to the T1DM group. The results of our study were consistent with those of the studies of Liu and Fany (5,19), who more clearly showed a regulating function disorder in the islet alpha cells in an FT1DM group. Sayama and cols. showed through morphological observations that islet alpha and beta cells were destroyed in FT1DM patients, while islet beta cells were destroyed and islet alpha cells remained functional in T1DM patients at the early stage of the disease (20). Fan and cols. followed 6 FT1DM patients for 9-72 months and reported that fasting and postprandial C-P2hr levels were close to levels requiring hospitalization at the acute stage of the disease, which suggested that the islet beta cells were completely and irreversibly destroyed (19). A report by Huang and cols. showed that the function of the islet beta cells in 2 FT1DM patients did not improve after 7 months of follow up (21). There were no significant differences in white blood cells, neutrophils, alanine, aspartate, potassium, cholesterol, high density lipoprotein, creatinine, and glutamine transpeptidase levels between the two groups. However, cholesterol and high-density lipoprotein levels were lower and white blood cells, neutrophils, alanine, aspartate, potassium, creatinine, and glutamine transpeptidase levels were generally higher in FT1DM patients at the acute stage [2]. This suggests that FT1DM caused not only metabolic and biochemical disorders but also tissue dysfunction or injuries, including in the liver and kidney, at the acute stage. Though metabolic and some biochemical indicators could return to the normal range after treatment at the stable stage, the liver and kidney injuries are irreversible, as indicated by the BUN and alkaline phosphatase levels being significantly higher in the FT1DM group compared to the T1DM group (P < 0.05). BUN is an important indicator of renal function, while alkaline phosphatase is mainly derived from the liver, bone, and kidneys; it indicates early kidney damage (22-24). Compromised kidney function following the acute stage of FT1DM may be caused by large blood glucose fluctuations, immune reactions, inflammation, and severe hypoglycemia. In conclusion, the present study suggests that patients with FT1DM at the stable stage and patients 589

Copyright© AE&M all rights reserved.

Blood glucose fluctuations in T1DM


Blood glucose fluctuations in T1DM

with T1DM had similar blood glucose fluctuation ranges. However, patients with FT1DM had a higher proportion of hypoglycemic levels, severely damaged pancreatic beta cells and more severe dysfunction of the islet alpha cells and kidney compared to patients with T1DM. Compliance with Ethical Standards: all procedures performed in studies involving human participants were completed in accordance with the ethical standards of the institutional and/or national research committee and the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Acknowledgements: we thank all patients and control subjects for participating in this study. Funding: this study was funded by the National Natural Science Foundation of China (NO. 81500606), the Jiangsu Provincial Department of Science and Technology Project (No. BL2014010) and the National Science Foundation of Jiangsu Province (No. SBK2015042970). Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Imagawa A, Hanafusa T, Miyagawa J, Matsuzawa Y. A novel subtype of type 1 diabetes mellitus characterized by a rapid onset and an absence of diabetes-related antibodies. Osaka IDDM Study Group. N Engl J Med. 2000;342(5):301-7. 2. Su XF, Fu LY, Wu JD, Xu XH, Li HQ, Sun R, et al. Fulminant type 1 diabetes mellitus: a study of nine cases. Diabetes Technol Ther. 2012;14(4):325-9. 3. Zheng C, Zhou Z, Yang L, Lin J, Huang G, Li X, et al. Fulminant type 1 diabetes mellitus exhibits distinct clinical and autoimmunity features from classical type 1 diabetes mellitus in Chinese. Diabetes Metab Res Rev. 2011;27(1):70-8. 4. Cho YM, Kim JT, Ko KS, Koo BK, Yang SW, Park MH, et al. Fulminant type 1 diabetes in Korea: high prevalence among patients with adult-onset type 1 diabetes. Diabetologia. 2007;50(11):2276-9. 5. Liu L, Mao J, Lu Z, Yan X, Ye Y, Jiang F. Clinical analysis of fulminant type 1 diabetes in China and comparison with a nationwide survey in Japan. Annales d’endocrinologie. 2013;74:36-9. 6. Imagawa A, Hanafusa T. Fulminant type 1 diabetes--an important subtype in East Asia. Diabetes Metab Res Rev. 2011;27(8):959-64. 7.

Imagawa A, Hanafusa T, Awata T, Ikegami H, Uchigata Y, Osawa H, et al. Report of the Committee of the Japan Diabetes Society on the Research of Fulminant and Acute-onset Type 1 Diabetes Mellitus: New diagnostic criteria of fulminant type 1 diabetes mellitus (2012). J Diabetes Investig. 2012;3(6):536-9.

9. Murase Y, Imagawa A, Hanafusa T, Iwahashi H, Uchigata Y, Kanatsuka A, et al. Fulminant type 1 diabetes as a high risk group for diabetic microangiopathy--a nationwide 5-year-study in Japan. Diabetologia. 2007;50(3):531-7. 10. Zhou J, Lv X, Mu Y, Wang X, Li J, Zhang X, et al. The accuracy and efficacy of real-time continuous glucose monitoring sensor in Chinese diabetes patients: a multicenter study. Diabetes Technol Ther. 2012;14(8):710-8. 11. Imagawa A, Hanafusa T, Uchigata Y, Kanatsuka A, Kawasaki E, Kobayashi T, et al. Fulminant type 1 diabetes: a nationwide survey in Japan. Diabetes Care. 2003;26(8):2345-52. 12. Su XF, Wu JD, Li Q, et al. Clinical features of 6 patients with fulminant type 1 diabetes mellitus. Chinese Journal of Diabetes Mellitus. 2010;2(5):329-33. 13. Luo S, Zhang Z, Li X, Yang L, Lin J, Yan X, et al. Fulminant type 1 diabetes: a collaborative clinical cases investigation in China. Acta Diabetol. 2013;50(1):53-9. 14. Kim HJ, Kim HS, Hahm JR, Jung JH, Kim SK, Lee SM, et al. The first Vietnamese patient with fulminant type 1 diabetes mellitus. Intern Med. 2012;51(17):2361-3. 15. Tsutsumi C, Imagawa A, Ikegami H, Makino H, Kobayashi T, Hanafusa T; Japan Diabetes Society Committee on Type 1 Diabetes Mellitus Research. Class II HLA genotype in fulminant type 1 diabetes: A nationwide survey with reference to glutamic acid decarboxylase antibodies. J Diabetes Investig. 2012;3(1):62-9. 16. Shibasaki S, Imagawa A, Hanafusa T. Fulminant type 1 diabetes mellitus: a new class of type 1 diabetes. Adv Exp Med Biol. 2012;771:20-3. 17. Aida K, Nishida Y, Tanaka S, et al. RIG-I- and MDA5-initiated innate immunity linked with adaptive immunity accelerates betacell death in fulminant type 1 diabetes. Diabetes. 2011;60:884-9. 18. Tanaka S, Nishida Y, Aida K, Maruyama T, Shimada A, Suzuki M, et al. Enterovirus infection, CXC chemokine ligand 10 (CXCL10), and CXCR3 circuit: a mechanism of accelerated beta-cell failure in fulminant type 1 diabetes. Diabetes. 2009;58(10):2285-91. 19. Fany YJ, Lu B, Yang JL. clinical analysis of six cases of fulminant type 1 diabetes mellitus. Chin J Diabetes. 2014;22(5):542-4. 20. Sayama K, Imagawa A, Okita K, Uno S, Moriwaki M, Kozawa J, et al. Pancreatic beta and alpha cells are both decreased in patients with fulminant type 1 diabetes: a morphometrical assessment. Diabetologia. 2005;48(8):1560-4. 21. Huang HB, Gong XY, Lin LX, Chen G. Approach to the patient with fulminant type l diabetes. Chin J Endocrinol Metab. 2014;30(1): 83-6. 22. Tuo SX, Qian YX, Yang L, Zhang YQ, Yu SQ. Relationship between Bone Mineral Density and Serum Bone Metabolism Biomarkers in Patients with Chronic Kidney Disease. Chin Med J. 2013;16(10):3422-4. 23. Li YP, Liao YH, Pan L, Ding WJ. Bone - kidney - parathyroid Axes Related Cytokines and Its Influencing Factors in Patients with Chronic Kidney Disease. 2014;19:2224-7. 24. Feng Q, Deng DY, Yuan WL, et al. Analysis of serum bone metabolism biochemical index of the Chronic Kidney Disease (CKD) 4-5 patients between diabetic nephropathy and non-diabetic nephropathy. Int J Lab Med. 2012;33(24):2992-3.

CopyrightŠ AE&M all rights reserved.

8. Koga M, Shimizu I, Murai J, Saito H, Kasayama S, Kobayashi T, et al. The glycated albumin to HbA1c ratio is elevated in patients

with fulminant type 1 diabetes mellitus with onset during pregnancy. J Med Invest. 2013;60(1-2):41-5.

590

Arch Endocrinol Metab. 2018;62/6


original article

Heart rate response to graded exercise test of elderly subjects in different ranges of TSH levels Rafael Cavalcante Carvalho1, Patrícia dos Santos Vigário2, Dhiãnah Santini de Oliveira Chachamovitz3, Diego Henrique da Silva Silvestre1, Pablo Rodrigo de Oliveira Silva2, Mario Vaisman4, Patrícia de Fátima dos Santos Teixeira4

ABSTRACT Objective: Life expectancy is increasing worldwide and studies have been demonstrating that elevated serum thyroid stimulating hormone (TSH) concentration in elderly is associated with some better health outcomes. This elevation is somewhat physiological as aging. The aim of this study was to investigate the heart rate (HR) response during a graded exercise test and its recovery in healthy elderly, comparing subjects within serum TSH in the lower limit of reference range to those within the TSH in the upper limit. Subjects and methods: A cross-sectional study was conducted with 86 healthy elderly aged 71.5 ± 5.1 years, with serum TSH between 0.4 – 4.0 mUl/mL. The participants were divided into two groups according to TSH level: < 1.0 mUl/mL (n = 13) and ≥ 1.0 µUI/mL (n = 73). All participants performed an ergometric test on a treadmill. The HR was recorded and analyzed at rest, during exercise and during the three minutes immediately after exercise. Results: No differences were observed in relation to HR at peak of exercise (TSH < 1.0 µUI/mL: 133.9 ± 22.5 bpm vs. TSH ≥ 1.0 µUI/mL: 132.4 ± 21.3 bpm; p = 0.70) and during the first minute of recovery phase (TSH < 1.0 µUI/ mL: 122.3 ± 23.1 bpm vs. TSH ≥ 1.0 µUI/mL: 115.7 ± 18.4 bpm p = 0.33). The groups also presented similar chronotropic index (TSH < 1.0 µUI/mL: 78.1 ± 30.6 vs. TSH ≥ 1.0 µUI/mL: 79.5 ± 26.4; p = 0.74). Conclusion: In this sample studied, there were no difference between lower and upper TSH level concerning HR response during rest, peak of exercise and exercise recovery. Arch Endocrinol Metab. Keywords Aged; thyroid hormones; heart rate; exercise

INTRODUCTION

L

ife expectancy is increasing worldwide (1), and studies focusing on the elderly population have been demonstrating that there is a positive correlation between aging and serum thyroid stimulating hormone (TSH) concentrations (2-4). For this population, higher serum TSH levels have been associated with better outcomes regarding mortality (3,5) and functionality (5-7) than those with serum TSH in the traditional “reference range.” The evidence of possible changes in the reference range for serum TSH in the elderly may be relevant not only to the upper but also the lower limit (8). It raises the idea that in the group of euthyroid elderly subjects, there might be some people with clinical signs and symptoms of subclinical hyperthyroidism (SCH), particularly in those ones with TSH near the low reference limit. Arch Endocrinol Metab. 2018;62/6

Correspondence to: Patrícia dos Santos Vigário Programa de Pós-Graduação em Ciências da Reabilitação, Centro Universitário Augusto Motta (UNISUAM) Praça das Nações, 34, Bonsucesso, 22041-010 – Rio de Janeiro, RJ, Brasil patriciavigario@yahoo.com.br Received on May/12/2018 Accepted on Sept/25/2018 DOI: 10.20945/2359-3997000000083

The association between SCH and increased cardiovascular risk and mortality is well established (9-11). Autonomic system alterations as vagal attenuation and sympathetic exacerbation are also described in SCH patients (12-15). Since abnormal heart rate (HR) behavior during exercise and recovery is related to autonomic dysfunction (16), cardiovascular risk and mortality (17,18), it would be adequate to study this parameter to indirectly predict the risk of cardiovascular events. Our group previously detected a high HR at rest, a low variation between HR at the peak of exercise and HR at rest and abnormal HR recovery (15) in middle-aged women with exogenous SCH. Considering that there is a trend towards higher values of TSH in the elderly (2-4) and that this condition is associated with better health-related variables in this population (5-8), it would be important 591

Copyright© AE&M all rights reserved.

2018;62(6):591-6

Laboratório de Ergoespirometria e Cineantropometria, Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brasil 2 Programa de Pós-Graduação em Ciências da Reabilitação, Centro Universitário Augusto Motta (UNISUAM), Rio de Janeiro, RJ, Brasil 3 Universidade Estácio de Sá, Rio de Janeiro, RJ, Brasil; Pesquisa Clínica Amil, CemedCare, Niterói, RJ, Brasil 4 Serviço de Endocrinologia, Hospital Universitário Clementino Fraga Filho (HUCFF), Rio de Janeiro, RJ, Brasil 1


Heart rate during exercise and TSH levels

to clarify if those with low levels of TSH would present cardiovascular impairments. Furthermore, since the elderly frequently suffer from cardiovascular disease (19), the HR variation during exercise and recovery are relevant outcomes that should be investigated. Therefore, the aim of this study was to examine the HR response during a graded exercise test and recovery in healthy elderly people with serum TSH within the reference range and compare subjects with serum TSH in the lower limit of reference to those with serum TSH in the upper limit.

SUBJECTS AND METHODS

Copyright© AE&M all rights reserved.

Study and sample Healthy elderly subjects of both genders, aged 65 or more years old without known thyroid dysfunctions and with serum TSH in the normal reference range (0.4–4.0 mUl/mL) were included in this crosssectional study. The exclusion criteria were: individuals who were athletes (defined as regular physical activity > 150 minutes/week, higher than 10 METs of intensity), any previous thyroid disease, hypothalamic-pituitary disease, congestive heart failure, chronic renal disease, acute respiratory disease, diabetes mellitus, stroke, chronic obstructive pulmonary disease, asthma, cancer, cirrhosis, aortic disease, pulmonary disease, peripheral vascular disease, and coronary artery disease. Subjects using corticosteroids, amiodarone, dopamine, or any drug that interferes with the levels of thyroid hormones and/or thyroid function were excluded. Subjects taking drugs that could interfere with autonomic adjustment mechanisms (beta blockers, calcium-channel blockers, and levodopa) were also excluded. Moreover, subjects who were bedridden, wheelchair users, and those who had any physical limitation that could affect the graded exercise testing were excluded. We divided the into two groups according to serum TSH levels: the first group was composed of those with serum TSH between 0.4 and 0.9 µUI/mL (lower reference range) and the second group included those with serum TSH between 1.0 and 4.0 µUI/mL. This cutoff point was chosen by considering cohort studies with the elderly population that showed adverse health effects in elderly people with TSH < 1.0 µUI/mL (20,21). The study was conducted at the Clementino Fraga Filho University Hospital (HUCFF) of the Federal 592

University of Rio de Janeiro, Brazil. The local ethics committee approved the study protocol (040/11CEP) in accordance with the ethical standards of the institutional and national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. All participants provided written consent before being enrolled in the study.

Laboratory analysis and exercise testing A physician obtained anamneses, performed physical examinations, measured vital signs, and reviewed the subjects’ medical records. Then, a fasting blood collection for laboratory analysis of serum TSH and free thyroxine (FT4) levels was scheduled within one week. TSH and free thyroxine (FT4) were measured with an immunochemiluminescence assay (Immulite; Diagnostic Products Corporation, Los Angeles, CA) with normal ranges of 0.4–4.0 µUI/mL and 0.8–1.9 ng/dL, respectively, at the Laboratory of Clinical Pathology/Hormone Section of HUCFF. The graded exercise testing was performed on a treadmill using a ramp protocol (22). Electrocardiogram CM5 derivation was utilized according to the Brazilian’s Ergometric National Consensus (23) using electrodes silver/silver chloride (Ag/AgCl). Participants were continuously monitored before the test (rest), during exercise, and during the first three minutes of exercise recovery. Systolic and diastolic blood pressure were checked before the test, every three minutes during exercise, and in the first and third minutes of exercise recovery (mercury sphygmomanometer; Narcosul 1400-C).The HR parameters were analyzed at three moments: when the subjects’ were at rest, during exercise, and during recovery immediately after the peak of exercise. They were considered: I) HR at rest (HRrest; bpm); II) peak HR (HRpeak; bpm); III) HR in the first and third minutes of recovery (HR1st min rec and HR 3rd min rec, respectively; bpm); IV) the difference between HRpeak and HRrest (∆HRpeak − HR rest; bpm); V) the difference between HRpeak and HR1st min (∆ HRpeak − HR1st min rec; bpm). A first-minute HR recovery ≤ 12 bpm was considered abnormal (17,24); VI) the difference between HRpeak and HR3rd min (∆ HRpeak − HR3rd min rec; bpm); and VII) chronotropic incompetence (CI). The CI was defined as the failure to get ≥ 80% of the predicted HR reserve (CI = (HRpeak – HRrest)/((220-age) − Arch Endocrinol Metab. 2018;62/6


Heart rate during exercise and TSH levels

HRrest) x 100) (25). The participants were stimulated to perform maximal exercise. Test interruption criteria were based on the American College of Sports and Medicine recommendations (23,26). The recovery was performed at 40% of the maximum velocity at peak exercise and 0% grade.

Statistical analyses All analyses were performed using the software SPSS 13.0 for Windows (SPSS, Inc.). Continuous variables were presented as the mean ± standard deviation (median) and categorical variables as the relative frequency. Distribution of the variables was assessed by the Kolmogorov-Smirnov test and presented nonparametric distribution. Comparisons between the groups according to TSH stratification (TSH < 1.0 µUI/mL vs. TSH ≥ 1.0 µUI/mL) were made using the Mann–Whitney U-test for continuous variables or the Chi-square test for categorical variables. To analyze the correlation between TSH (as a continuous variable) and HR parameters at rest, at peak of exercise and during recovery after exercise, the logarithmic transformation of TSH was made. After the certification that the logarithm of TSH assumed a normal distribution through Kolmogorov-Smirnov test, the Pearson’s

correlation coefficient was calculated. The statistical significance was set at p < 0.05.

RESULTS A total of 86 participants were included in the study. Most of them were female (67.4%) and had serum TSH ≥ 1.0 µUI/mL (84.9%). The hormonal and clinical characteristics of the sample are shown in Table 1. Elderly participants with TSH < 1.0 µUI/mL and TSH ≥ 1.0 µUI/mL did not differ in terms of age, body mass index (BMI), gender, or FT4 (p > 0.05). The HR parameters we considered in the study are presented in Table 2. No differences were observed between elderly patients with serum TSH < 1.0 µUI/mL and TSH ≥ 1.0 µUI/mL in relation to HR during rest, at peak exercise and during recovery after exercise (all p-values > 0.05). Concerning the first minute of recovery after exercise, 46.2% of elderly patients with TSH < 1.0 µUI/mL and 31.5% of elderly patients with TSH ≥ 1.0 µUI/mL had an abnormal HRpeak recovery i.e., less than or equal to 12 bpm, with no statistical difference between the groups (p = 0.35). The mean chronotropic index of the whole group (n = 86) was 79.3 ± 26.9 (79.1)% and there were

Table 1. Clinical and hormonal characteristics of elderly patients that participated in the study Whole group (n = 86)

TSH < 1.0 µUI/mL (n = 13)

TSH ≥ 1.0 µUI/mL (n = 73)

p-value*

Age (years)

71.5 ± 5.1 (70.5)

71.8 ± 5.1 (71.5)

71.4 ± 5.2 (70.0)

0.54

BMI (kg/m )

27.3 ± 4.4 (26.3)

28.9 ± 4.7 (27.2)

27.2 ± 4.4 (26.3)

0.58

67.4%

53.8%

69.9%

0.22†

TSH (µUI/mL)

1.9 ± 1.1 (1.6)

0.7 ± 0.1 (0.7)

2.1 ± 1.1 (1.7)

0.00*

FT4 (ng/dL)

1.1 ± 0.2 (1.1)

1.2 ± 0.2 (1.1)

1.1 ± 0.6 (1.0)

0.08*

2

Gender (female; %)

Results are presented as mean ± standard-deviation (median) for continuous variables and as (%) for categorical variables; *Mann-Whitney U-test for numeric and (†) Fisher’s Exact test for categorical variables; TSH < 1.0 µUI/mL vs. TSH ≥ 1.0 µUI/mL; statistical significance set at p < 0.05; BMI: body mass index; TSH: thyroid stimulating hormone; FT4: free thyroxine.

Whole group (n = 86)

TSH < 1.0 µUI/mL (n = 13)

TSH ≥ 1.0 µUI/mL (n = 73)

p-value*

HR rest (bpm)

74.7 ± 13.8 (73.5)

77.3 ± 13.1 (74.0)

74.3 ± 13.9 (73.0)

0.73

HR peak (bpm)

132.6 ± 21.4 (133.5)

133.9 ± 22.5 (133.0)

132.4 ± 21.3 (134.0)

0.70

57.8 ± 21.1 (54.5)

56.5 ± 20.5 (59.0)

58.1 ± 21.4 (54.0)

0.56

116.7 ± 19.2 (116.5)

122.3 ± 23.1 (122.0)

115.7 ± 18.4 (116.0)

0.33

Δ HR peak-rest (bpm) HR 1 min (bpm) st

Δ HR peak- 1 min rec (bpm) st

HR 3rd min (bpm) Δ HR peak- 3 min rec (bpm) rd

15.8 ± 11.2 (16.5)

11.5 ± 14.3 (16.0)

16.6 ± 10.5 (17.0)

0.33

104.8 ± 17.1 (104.5)

110.0 ± 20.6 (107.0)

103.8 ± 16.4 (104.0)

0.33

11.9 ± 8.5 (11.0)

12.3 ± 8.4 (12.0)

11.8 ± 8.6 (11.0)

0.87

Results are presented as mean ± standard-deviation (median); *Mann-Whitney U-test; TSH < 1.0 µUI/mL vs. TSH ≥ 1.0 µUI/mL; statistical significance set at p < 0.05; TSH: thyroid stimulating hormone HR: heart rate; Δ HR: heart rate variation; min: minute; rec: recovery. Arch Endocrinol Metab. 2018;62/6

593

Copyright© AE&M all rights reserved.

Table 2. Heart rate parameters at rest, at peak of exercise and during recovery after exercise, according to TSH stratification


Heart rate during exercise and TSH levels

no differences according to TSH stratification [TSH < 1.0 µUI/mL = 78.1 ± 30.6 (76.6)% vs. TSH ≥ 1.0 µUI/mL = 79.5 ± 26.4 (79.6)%; p = 0.74]. No correlations were found between the logarithm of TSH and HR parameters at rest, at peak exercise and during recovery after exercise. The results are present in Table 3. Table 3. Correlation between logarithm of TSH and heart rate parameters at rest, at peak of exercise and during recovery after exercise Logarithm of TSH Correlation coefficient

p-value*

HR rest (bpm)

-0.010

0.465

HR peak (bpm)

-0.020

0.429

Δ HR peak-rest

-0.014

0.451

HR 1 min

-0.052

0.319

Δ HR peak- 1st min rec

0.051

0.323

st

* Pearson’s test; statistical significance set at p < 0.05; TSH: thyroid stimulating hormone; HR: heart rate; Δ HR: heart rate variation; min: minute; rec: recovery.

Copyright© AE&M all rights reserved.

DISCUSSION To our knowledge, this is the first study that has investigated the relationship between TSH variation in the normal range and HR parameters in healthy elderly subjects. The main findings were that different serum TSH levels in the normal reference range did not associate with HR abnormalities during rest, exercise, or recovery. High values of HR at rest are associated with impaired functioning of the autonomic nervous system (ANS), with exacerbation of the sympathetic nerve and decreased vagal modulation (27-29), and this can mean less variation in the difference between HRpeak and HRrest (∆HRpeak − HR rest). The variability of HR is an important mark of chronotropic incompetence (CI), defined as the failure to get ≥ 80% of the HR reserve and described as a predictor of cardiovascular mortality and CHD events (24,30). In the population, with exogenous subclinical hyperthyroidism, our group previously demonstrated values of HR compatible with CI in middle-aged women and increased resting HR (15). The CI observed in our study, however, did not demonstrate a relationship with TSH. This may be because in an elderly population there are indications of decline in parasympathetic stimulus with a decrease in HR variability (31). Another interesting finding, reported by Magrì and cols. (32) about the difference between HRpeak and 594

HRrest (∆HRpeak − HRrest) compatible with CI in patients with CHF, was the correlation with functional capacity and peak oxygen uptake, i.e., the smaller the ∆HRpeak–HRrest, the smaller the functional capacity classified by the New York Heart Association. It is noteworthy that despite these findings, the authors warned to not consider a cause/effect relationship. Furthermore, Vigário and cols. (15) reported CI and impaired exercise capacity with lower oxygen consumption in middle-aged women, with exogenous subclinical hyperthyroidism; however, in our study, despite the fact that the CI could be a marker of impaired exercise capacity, it did not show a correlation with TSH near the low limit of the normal reference range. Looking for association of TSH and functional capacity in elderly people with preexisting cardiovascular disease or who are at risk to develop such a condition, Virgini and cols. (33) correlated this with quartiles of TSH. They evaluated 5,182 participants with a mean of age 75.3 years. The groups were similar for most variables, except for BMI (lower in SCH) and hypertension (higher in subclinical hypothyroidism). Functional capacity was measured using two questionnaires: the Barthel Index (a questionnaire used to assess self-care activities of daily living and an individual’s level of dependence) and the Instrumental Activities of Daily Living (a questionnaire that also measures activities of daily living, but involves interaction with the physical and social environment). No association between TSH and functional capacity were observed. In addition, after a 3-year cohort, a decrease in functional capacity without association with TSH was observed. Our results are in accordance with and provide evidence for the relevance of TSH into the normal reference range as a single mark for a decline of functional capacity. Concerning the recovery from exercise, the first minute is an important mark of vagal imbalance, in other words, it is difficult to achieve at least 12 bpm of ∆ HRpeak − HR1st min recovery difference. This abnormality is associated with an altered function in ANS, also described with the predictor of cardiovascular risk and all-cause mortality, including the elderly people (17,23). In the population, with subclinical hyperthyroidism, studies demonstrated impaired functioning of ANS (8,12,14,34); however, the patients who presented that difficult, rapidly controlled HR during recovery of exercise, were not mainly for any of the two TSH groups in our sample. Arch Endocrinol Metab. 2018;62/6


Heart rate during exercise and TSH levels

Arch Endocrinol Metab. 2018;62/6

≥ 1.0 = 73 participants) do not affect the Mann–Whitney test properties, we recognize that the statistical power of the test diminishes as the group size becomes more unequal (39). Future analysis, including larger sample sizes of both groups, should be useful for gaining a better understating of the associations between TSH levels and HR in the elderly population. Another limitation of our study is the absence of division groups in sedentary and physically active people. In conclusion, the results of this study did not show a relationship between TSH and HR impairments during rest, at peak exercise, or during exercise recovery in elderly people. The groups with different levels of TSH showed a similar behavior for all variables considered in the study. Further investigations with older people (> 80 years) are necessary, whereas this population presents a higher level of TSH than our sample and it can provide a different outcome. Acknowledgments: National Council of Technological and Scientific Development (CNPq); José Bonifácio University Foundation (FUJB); Carlos Chagas Filho Research Support Foundation (FAPERJ). Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. World Health Organization (WHO). National Institute on Aging, National Institutes of Health and U.S. Department of Health and Human Services: Global health and aging; 2011. 2. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, et al. Serum TSH, T4 and Thyroid antibodies in the United States population (1988-1994): National Health and Nutrition Examination Survey (NHANESII). J Clin Endocrinol Metab. 2002;87(2):489-99. 3. Atzmon G, Barzilai N, Hollowell JG, Surks MI, Gabriely I. Extreme longevity is associated with increased serum thyrotropin. J Clin Endocrinol Metab. 2009;94(4):1251-4. 4. Bremner AP, Feddema P, Leedman PJ, Brown SJ, Beilby JP, Lim EM, et al. Age-related changes in thyroid function: a longitudinal study of a community-based cohort. J Clin Endocrinol Metab. 2012; 97(5):1554-62. 5. Gussekloo J, van Exel E, de Craen AJ, Meinders AE, Frölich M, Westendorp RG. Thyroid status, disability and cognitive function, and survival in old age. JAMA. 2004; 292(21):2591-9. 6. Simonsick EM, Newman AB, Ferrucci L, Satterfield S, Harris TB, Rodondi N, et al. Subclinical hypothyroidism and functional mobility in older adults. Arch Intern Med. 2009;169(21): 2011-7. 7. Silva SO, Chan IT, Santos LMA, Cohen M, Araújo MLRP, Almeida JS, et al. Impact of thyroid status and age on comprehensive geriatric assessment. Endocrine. 2013;47(1):255-65. 8. Goichot B, Sapin R, Schlienger JL. Subclinical Hyperthyroidism: considerations in defining the lower limit of the thyrotropin reference interval. Clin Chem. 2009;55(3):420-4. 9. Sgarbi JA, Matsumura LK, Kasamatsu TS, Ferreira SR, Maciel RMB. Subclinical thyroid dysfunctions are independent risk

595

Copyright© AE&M all rights reserved.

Recent studies have raised the question of whether the reference limits of TSH should be reconsidered in the elderly (8,35). Elderly patients with slightly elevated serum TSH were previously categorized as subclinical hypothyroidism patients; however, nowadays they may be classified as euthyroid subjects. In addition, previous studies showed slight benefits in the elderly from a mild increase in serum TSH, even when compared with euthyroid subjects (5). But we did not find impairment of health associated with serum TSH near the low reference range, i.e., between 0.4 and 0.9 µUI/mL, and according to Fontes and cols., (36) the low reference range remains the same in different age groups with changes in the upper limit. Collet and cols., (37) observed that the higher risk of atrial fibrillation, coronary heart disease mortality, and total mortality in patients with subclinical hyperthyroidism is mainly when the serum TSH level was lower than 0.10 mIU/L on the study with 10 cohorts. We considered the TSH cut-off point to be < 1.00 µUI/mL and ≥ 1.00 µUI/mL in view of cohort studies with elderly people that reported adverse health effects in those with serum TSH levels < 1.00 µUI/mL (20,21). Studies have been demonstrated that serum TSH levels below 1.0 µUI/mL, even in the reference range, may have negative outcomes in elderly subjects concerning depressive symptoms and risk of hip fracture (20,21). By the possible correspondence with adverse effects related to subclinical hyperthyroidism with HR impairments, the study of TSH range next to those low abnormal values seems to be justified. Our group evaluated the effect of antithyroid drug use in order to increase serum TSH levels to the upper normal range in elderly subjects with initial TSH levels < 1.0 μIU/mL, and analyzed the cardio-respiratory effort outcomes in a cross-sectional cohort. The results did not corroborate the hypothesis that lower serum TSH levels within the normal reference range had a negative impact on quality of life or cardiopulmonary capacity during exercise in healthy elderly people. Furthermore, the intervention intending to increase serum TSH to the upper normal range did not generate significant changes in the evaluated outcomes (38). Admitting the difficult-to-get volunteers considering the inclusion and exclusion criteria we adopted, our small sample size – mainly those who compose the group with TSH levels < 1.0 µUI/mL – is the major limitation of the study. Although the differences in the sample sizes (i.e., TSH < 1.0 = 13 participants and TSH


Heart rate during exercise and TSH levels

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

factors for mortality in a 7.5-year follow-up: the Japanese– Brazilian thyroid study. Eur J Endocrinol. 2010;162(3):569-77. Gencer B, Collet T, Virgini V, Auer R, Rodondi N. Subclinical thyroid dysfunction and cardiovascular outcomes among prospective cohort studies. Endocr Metab Immune Disord Drug Targets. 2013;13(1):4-12. Aggarwal N, Razvi S. Thyroid and aging or the aging thyroid? An evidence-based analysis of the literature. J Thyroid Res. 2013;2013:1-8. Petretta M, Bonaduce D, Spinelli L, Vicario MLE, Vincenzo N, Marciano F, et al. Cardiovascular hemodynamics and cardiac autonomic control in patients with subclinical and overt hyperthyroidism. Eur J Endocrinol. 2001;145(6):691-6. Osman F, Franklyn J, Daykin J, Chowdhary S, Holder RL, Sheppard MC, et al. Heart rate variability and turbulence in hyperthyroidism before, during, and after treatment. Am J Cardiol. 2004;94(4):465-9. Portella RB, Pedrosa RC, Coeli CM, Buescu A, Vaisman M. Altered cardiovascular vagal responses in nonelderly female patients with subclinical hyperthyroidism and no apparent cardiovascular disease. Clin Endocrinol (Oxf). 2007;67(2):290-4. Vigário PS, Chachamovitz DS, Teixeira PF, Santos MA, Oliveira FP, Vaisman M. Impaired functional and hemodynamic response to graded exercise testing and its recovery in patients with subclinical hyperthyroidism. Arq Bras Endocrinol Metabol. 2011;55(3):203-12. Evrengul H, Tanriverdi H, Kose S, Amasyali B, Kilic A, Celik T, et al. The relationship between heart rate recovery and heart rate variability in coronary artery disease. Ann Noninvasive Electrocardiol. 2006;11(2):154-62. Cole CR, Blackstone EH, Pashkow FJ, Snader CE, Lauer MS. Heart-rate recovery immediately after exercise as a predictor of mortality. N Engl J Med. 1999;28:341(18):1351-7. Thayer JF, Lane RD. The role of vagal function in the risk for cardiovascular disease and mortality. Biol Psychol. 2007;74(2): 224-42. Dai DF, Chen T, Johnson SC, Szeto H, Rabinovitch PS. Cardiac aging: from molecular mechanisms to significance in human health and disease. Antioxid Redox Signal. 2012;16(12):1492-526. Medici M, Direk N, Visser WE, Korevaar TI, Hofman A, Visser TJ, et al. Thyroid function within the normal range and the risk of depression: a population-based cohort study. J Clin Endocrinol Metab. 2014;99(4):1213-9. Leader A, Ayzenfeld RH, Lishner M, Cohen E, Segev D, Hermoni D. Thyrotropin levels within the lower normal range are associated with an increased risk of hip fractures in euthyroid women, but not men, over the age of 65 years. J Clin Endocrinol Metab. 2014;99(8):2665-73. Maeder M, Wolber T, Atefy R, Gadza M, Ammann P, Myers J, et al. A nomogram to select the optimal treadmill ramp protocol in subjects with high exercise capacity: validation and comparison with the Bruce protocol. J Cardiopulm Rehabil. 2006;26(1):16-23. Mastrocolla LE, Brito AX, Brito FS, Castro I, Godoy M, Alfieri RG. Consenso Nacional de ergometria. Arq Bras Cardiol. 1995;65(2):189-211.

25. Lauer MS, Francis GS, Okin PM, Pashkow FJ, Snader CE, Marwick TH. Impaired chronotropic response to exercise stress testing as a predictor of mortality. JAMA.1999;281(6):524-9. 26. American College of Sports Medicine (ACSM). Guidelines of exercise testing and exercise prescription. 8th ed. Philadelphia: Lea & Febiger, 2009. 27. Chen JL, Chiu HW, Tseng YJ, Chu WC. Hyperthyroidism is characterized by both increased sympathetic and decreased vagal modulation of heart rate: evidence from spectral analysis of heart rate variability. Clin Endocrinol. 2006;64(6):611-6. 28. Galloe AM, Rolff M, Nordin H, Ladefoged SD, Mogensen NB. Cardiac performance and thyroid function. The correlation between systolic time intervals, heart rate and thyroid hormone levels. Dan Med Bull. 1993;40(4):492-5. 29. Karthik S, Pal GK, Nanda N, Hamide A, Bobby Z, Amudharaj et al. Sympathovagal imbalance in thyroid dysfunctions in females: Correlation with thyroid profile, heart rate and blood pressure. Indian J Physiol Pharmacol. 2009;53(3):243-52. 30. Girotra S, Kitzman DW, Kop WJ, Stein PK, Gottdiener JS, Mukamal KJ. Heart rate response to a timed walk and cardiovascular outcomes in older adults: the cardiovascular health study. Cardiology. 2012;122(2):69-75. 31. Kaye DM, Esler MD. Autonomic control of the aging heart. Neuro Molecular Med. 2008;10(3):179-86. 32. Magrì D, Palermo P, Cauti FM, Contini M, Farina S, Cattadori G, et al. Chronotropic incompetence and functional capacity in chronic heart failure: no role of β‐blockers and β‐blocker dose. Cardiovasc Ther. 2012;30(2):100-8. 33. Virgini VS, Wijsman LW, Rodondi N, Bauer DC, Kearney PM, Gussekloo J, et al. Subclinical thyroid dysfunction and functional capacity among elderly. Thyroid. 2014;24(2):208-14. 34. Eustatia-Rutten CFA, Corssmit EPM, Heemstra KA, Smit JWA, Schoemaker RC, Romijn JA, et al. Autonomic nervous system function in chronic exogenous subclinical thyrotoxicosis and the effect of restoring euthyroidism. J Clin Endocrinol Metab. 2008;93(7):2835-41. 35. Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr Rev. 2008;29(1):76-131. 36. Fontes R, Coeli CR, Aguiar F, Vaisman M. Reference interval of thyroid stimulating hormone and free thyroxine in a reference population over 60 years old and in very old subjects (over 80 years): comparison to young subjects. Thyroid Res. 2013;6(1):1-8. 37. Collet TH, Gussekloo J, Bauer DC, Elzen WPJ, Cappola AR, Balmer P, et al. Subclinical hyperthyroidism and the risk of coronary heart disease and mortality. Arch Intern Med. 2012;172(10):799-809. 38. Chachamovitz DSO, Vigário PS, Carvalho RC, Silvestre DHS, Moerbeck AEV, Soffientini M, et al. Does low serum TSH within the normal range have negative impact on physical exercise capacity and quality of life of healthy elderly people? Arch Endocrinol Metab. 2015;60(3):236-45. 39. Mann HB, Whitney DR. On a test of whether one of two random variables is stochastically larger than the other. Ann Math Statist. 1947;18(1):50-60.

Copyright© AE&M all rights reserved.

24. Nishime EO, Cole CR, Blackstone EH, Pashkow FJ, Lauer MS. Heart rate recovery and treadmill exercise score as predictors

of mortality in patients referred for exercise ECG. JAMA. 2000;284(11):1392-8.

596

Arch Endocrinol Metab. 2018;62/6


original article

Risk factors for new-onset diabetes mellitus after kidney transplantation (NODAT): a Brazilian single center study Camila Lima1, Amanda Grden1, Thelma Skare1, Paulo Jaworski1, Renato Nisihara1,2

ABSTRACT

Keywords New-onset diabetes after transplantation (NODAT); diabetes mellitus; renal transplant; calcineurin inhibitors

INTRODUCTION

N

ew-onset diabetes after transplant, or NODAT, is one of the major complications after solid organ transplantation (1,2). There is a five- to six-fold higher incidence of new-onset diabetes mellitus (DM) in the first year post kidney transplantation in comparison to waiting-listed patients (3). This complication is considered to lead to graft failure and to promote cardiovascular disease, one of the main causes of death in transplant recipient (4). A large meta-analysis has shown that NODAT is present in 2%–50% of patients, 1 year after the transplant procedure (5). The pathophysiology of NODAT closely mimics type 2 DM: insulin resistance and insulin hyposecretion develop in both situations. Nevertheless, in NODAT, insulin hyposecretion seems to be a crucial determining factor in glucose tolerance deterioration (4). Arch Endocrinol Metab. 2018;62/6

Universidade Evangélica, Departamento de Medicina, Curitiba, PR, Brasil 2 Universidade Positivo, Departamento de Medicina, Curitiba, PR, Brasil 1

Correspondence to: Renato Nisihara Departamento de Medicina, Rua Padre Agostinho, 2770 80730-000 – Curitiba, PR, Brasil renatonisihara@gmail.com Received on Aug/5/2017 Accepted on Dec/13/2017 DOI: 10.20945/2359-3997000000084

Several risk factors were known to be associated with NODAT, such as ethnic background, family history of diabetes, sedentary lifestyle, high body mass index (BMI), and cytomegalovirus and hepatitis C virus (HCV) infection. A high dose of glucocorticoids and the use of immunosuppressive regimen with calcineurin inhibitors may also be associated with this condition (6-8). Numakura and cols. (9) found that genetic factors such as certain vitamin D receptor haplotypes lead to increased risk of NODAT. Kim and cols. (10) found that Toll-like receptor (TLR)-4 and TLR-6 gene polymorphisms were significantly associated with NODAT, showing that the innate immune system and inflammation via TLR activation may have an important role in the pathogenesis of this complication. The results of some studies indicate that several genes, such as interleukins, transcription factor 7-like 2, solute 597

Copyright© AE&M all rights reserved.

Objectives: This study aims to verify the new-onset diabetes after kidney transplant (NODAT) incidence in recipients within 1 year after kidney transplantation from a single center in Southern Brazil and to assess the associated conditions. Subjects and methods: A retrospective study of 258 postrenal transplant patients was performed. Demographic (gender, age, ethnic background) and clinical (origin of graft, associated infections, body mass index (BMI) at transplant time and 6 and 12 months after, causes of renal failure, and comorbidities) data were analyzed. All patients were on tacrolimus, mycophenolate mofetil, and prednisone treatment. Patients with and without NODAT were compared. Results: A NODAT incidence of 31.2% was noted 1 year post transplantation. In the univariate analysis, patients with NODAT were older (p = 0.001), mostly had African–American ethnic background (p = 0.02), and had renal failure secondary to high blood pressure (HBP) (p = 0.001). The group of patients with NODAT also had more incidence of post-transplant HBP (p = 0.01), heart failure (p = 0.02), and dyslipidemia (p = 0.001). Logistic regression showed that African–American ethnic background, posttransplant HBP, and dyslipidemia were independently associated with NODAT. Conclusion: This study shows a NODAT incidence that is greater in patients with African–American ethnic background and that is associated with HBP and dyslipidemia. Arch Endocrinol Metab. 2018;62(6):597-601


Diabetes after kidney transplantation

carrier family 30 (zinc transporter), member 8 genes (SLC30A8), matrix metalloproteinases (MMPs), and chemokine (C–C motif) ligand 5 (CCL5) genes, were related with the development of NODAT (11-15). Genetic background may influence the prevalence of NODAT in different populations (6). The increased survival of renal transplant patients requires adequate attention as its complication may interfere with patients’ survival and quality of life (16,17). Aiming to know the prevalence of NODAT and its risk factors in the region, a cohort of kidney transplant recipients from Southern Brazil were studied.

Copyright© AE&M all rights reserved.

SUBJECTS AND METHODS This retrospective study was approved by the local Committee of Ethics in Research (56934016.4.0000.0103). Charts of kidney transplant patients from a single center, a university evangelical hospital, located in the city of Curitiba, Paraná, Brazil, registered from July to December 2015, were reviewed for epidemiological, clinical, and treatment profile. Patients under 18 years of age, those with DM prior to transplantation, those who had first-degree relatives with DM, and those who had used immunobiological drugs or had graft rejection were excluded. The study sample included 258 patients (141/268 or 52.6% males, with median age of 43 years; range 18– 75 years). All patients were on standard treatment, with prednisone, mycophenolate mofetil, and tacrolimus, which is determined by the Brazilian Health Ministry, Ordinance 1.168, in 2004. The following data of all patients were reviewed retrospectively for 12 months: BMI prior to transplantation and six and twelve months post transplantation, causes of renal failure prior to transplantation, graft origin (either from deceased or living donor), and occurrence of hepatitis C and CMV infections. The diagnosis of DM was made according to the American Diabetes Association, through the analysis of three altered values of blood glucose, which was done by the transplant doctor and was registered in the patient medical record 12 months after transplantation (18). The obtained data was collected in frequency and contingency tables. The epidemiological and clinical profiles between patients with and without NODAT were compared using the Fisher exact test and chisquare tests for nominal data and unpaired t-test 598

and Mann–Whitney test for numeric data. To assess variables independently, logistic regression was used. The significance level adopted was 5%.

RESULTS The description of clinical, epidemiological, and treatment data of kidney transplant patients being studied are shown in Table 1. Only three patients had HCV infection. In this sample, the NODAT incidence was 31.2%. Table 2 shows the results obtained between patients with and without NODAT were compared. It is noticeable that elder patients, African–American background patients, and the group with hypertension and dyslipidemia had a higher NODAT incidence. Patients with NODAT also experience more hypertension and less chronic glomerulonephritis, which are causes of renal failure that required transplantation. When all variables with p < 0.1 were studied through logistic regression, it was found that African–American ethnic background (p = 0.03; OR = 2.08; 95% CI = 1.05-4.14), dyslipidemia (p = 0.03; OR = 2.08; 95% CI = 1.09–5.12), and hypertension (p = 0.04; OR = 4.7; 95% CI = 1.04–21.5) were independently associated with NODAT.

DISCUSSION In the present study, which included 258 renal post transplanted recipients, all in the same therapeutic regimen, there was a NODAT incidence of 31.2% in the first year after the transplant procedure. It is noteworthy that individuals with DM in the first-degree relatives in the sample were excluded, so the possible occurrence of a common type 2 DM was minimized. In addition, it was observed that NODAT was independently associated with factors such as African–American background, arterial hypertension, and dyslipidemia. Other authors have found that a myriad of factors affect the development of NODAT, such as high BMI, the use of calcineurin inhibitors and corticosteroids, old age, and cytomegalovirus (CMV) and hepatitis C infection (1). BMI alone was not linked to the appearance of this complication in the sample. However, the sample’s mean BMI ranged from 24 to 26 kg/m2 which is close to the accepted normal range. Therefore, obesity, which is widely linked to DM, was Arch Endocrinol Metab. 2018;62/6


Diabetes after kidney transplantation

Table 1. Clinical, epidemiological and treatment data of kidney transplanted patients (n = 258) Variable Ethnic background (auto declared)

Causes of renal failure

Graft type

Co-morbidities

n

(%) or central tendency

209

35;78.6

1

0.3

Afro descendants

53

20.9

Chronic glomerulonephritis

125

51.9

Hypertension

43

16.6

Polycystic kidney disease

21

8.1

Pyelonephritis and litiasis

12

4.6

IgA nephropathy

11

4.2

Others

37

14.6

Caucasians Asian

6 months post transplantation

24.2 (22.2-28.5)

12 months post transplantation

26.1 (23.1-29.7)

Cadaveric

121

46.8

Living donor (relative)

98

37.9

Living donor (not relative)

37

14.3

Unknown

2

0.7

Arterial hypertension

229

87.5

Dyslipidemia

38

14.3

Ischemic heart disease

10

3.8

Heart failure

3

1.1

BMI: body mass index; CMV: cytomegalovirus.

With NODAT 31.2%

Without NODAT 68.8%

P

Median age (years)

48 (37-56)

41 (26-51)

0.001

Ethnic background

Afro descend – 30.2% Caucasian – 69.7%

Afro descend – 17.5% Caucasian – 82.4%

0.02*

Male gender

62%

52.8%

0.17

Causes of renal failure

Glomerulonephritis – 35.8% Hypertension – 28.2% Others – 35.8%

Glomerulonephritis – 56.6% Hypertension – 12.0% Others – 31.3%

0.001**

Graft donor

Deceased – 53.1% Living related – 36.7% Living not related – 10.1%

Decesead – 44.7% Living related – 15.6% Living not related – 39.2%

0.34

24.9 (22.0-27.9)

24.5 (21.5-28.1)

0.49

Mean BMI (kg/m2) 6 months post-transplant

25.5 ± 4.1

25.4 ± 4.2

0.78

Mean BMI (kg/m2) 12 months post-transplant

26.4 ± 4.7

26.3 ± 4.5

0.89

Associated arterial hypertension

97.4%

85.6%

0.01#

Associated dyslipidemia

25.3%

9.7%

0.001##

Associated heart failure

3.7%

0

0.02§

5.06%

9.7%

0.50

1.2%

4.02%

0.44

Median BMI (kg/m2) pre transplant

CMV infection BMI: body mass index; CMV: cytomegalovirus.

#

OR: 4.3; 95% CI: 1.2-14.7. ## OR: 3.1 (1.4-6.3). § OR: 15.9; 95%

CI: 0.8-313.1. Arch Endocrinol Metab. 2018;62/6

599

Copyright© AE&M all rights reserved.

Table 2. Comparison of renal transplanted patients according to presence or not of NODAT (New Onset Diabetes After Transplantation)


Copyright© AE&M all rights reserved.

Diabetes after kidney transplantation

underrepresented in the sample. This may explain the diversity of the results found. Additionally, CMV infection could not be associated with NODAT in this study, considering the very low incidence of infection in patients. The same occurs with HCV infection. However, literature concerning HCV infection and NODAT is ambiguous, with some authors supporting higher prevalence of these findings (19) and others denying it (20,21). In this sample, two of three patients with HCV infection developed DM. NODAT’s association with African–American background was also described by other authors (19). This association was also observed in the Brazilian sample, regardless of the fact that Brazilian people have a highly mixed ethnic background. Tacrolimus is considered one of the main NODAT causes, (22) and all of the patients were receiving it. A calcineurin inhibitor has been considered the cornerstone of immunosuppressive regimens in renal transplant (22). It inhibits T-cell activation, thereby inhibiting interleukin-2 syntheses, and leads to a failure of T-cell clonal expansion (23). Several authors have shown that patients treated with tacrolimus have higher NODAT incidence when compared to those who were treated with cyclosporine (1,24). Nonetheless, tacrolimus was shown to be superior to cyclosporine in terms of patient mortality, graft loss, and hypertension development (24). The mechanism involved in tacrolimus-induced NODAT includes diminished insulin secretion, as calcineurin is expressed in pancreas β cells. This medication disrupts mitochondrial permeability, causes inhibition of NFAT transcription factor, and targets cAMP-responsive element-binding protein transcriptional coactivator (3). In addition, it inhibits glucose uptake from muscular cells and adipocytes (3). Hypomagnesemia is one of the proposed mechanisms for calcineurin-associated glucose intolerance, due to magnesium which is essential for glucose transport and pancreatic insulin secretion, and is involved in the postreceptor insulin signaling (25). NODAT is associated with lower survival of transplanted recipients, and it is an independent risk factor linked to graft failure (22). Furthermore, classic DM complications are seen in patients with NODAT such as peripheral neuropathy, ketoacidosis, hyperosmolar coma, and even biopsy-proven diabetic nephropathy (22,26). Serum creatinine levels are significantly higher in patients with NODAT compared to those without it after 5 years of transplantation 600

(22,26). All these complications lead to increased mortality, mainly due to cardiovascular events (22,26). Cosio and cols. (27) demonstrated that the 5-year cumulative incidence of cardiovascular events was correlated with fasting glucose levels seen 1 year after transplantation. It was found that a higher rate of hypertension and dyslipidemia in the NODAT sample is known to be a risk factor for atherosclerotic disease and cardiovascular events. This is a retrospective study, which is also considered one of the limitations. It is also possible that the occurrence of NODAT would be higher in longer periods of observation. In conclusion, this study shows a higher NODAT incidence in kidney transplant recipients with African– American ethnic background. Moreover, NODAT is associated with higher blood pressure levels and dyslipidemia in all transplant recipients, regardless of the ethnic background. Prospective and multicentric studies may be useful to better understand and prevent NODAT. Funding of sources: none. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Yu H, Kim H, Baek CH, Baek SD, Jeung S, Han DJ, et al. Risk factors for new-onset diabetes mellitus after living donor kidney transplantation in Korea – A retrospective single center study. BMC Nephrol. 2016;17(1):106. 2. Räkel A, Karelis AD. New-onset diabetes after transplantation: risk factors and clinical impact. Diabetes Metab. 2011;37(1):1-14. 3. Chakkera HA, Kudva Y, Kaplan B. Calcineurin Inhibitors: Pharmacologic mechanisms impacting both insulin resistance and insulin secretion leading to glucose dysregulation and diabetes mellitus. Clin Pharmacol Ther. 2017;101(1):114-20. 4. Davidson J, Wilkinson A, Dantal J, Dotta F, Haller H, Hernández D, et al. New-onset diabetes after transplantation: 2003 International consensus guidelines. Proceedings of an international expert panel meeting. Barcelona, Spain, 19 February 2003. Transplantation. 2003 May 27;75(10 Suppl):SS3-24. 5. Montori VM, Basu A, Erwin PJ, Velosa JA, Gabriel SE, Kudva YC. Posttransplantation diabetes: a systematic review of the literature. Diabetes Care. 2002;25(3):583-92. 6. Kurzawski M, Dziewanowski K, Łapczuk J, Wajda A, Droździk M. Analysis of common type 2 diabetes mellitus genetic risk factors in new-onset diabetes after transplantation in kidney transplant patients medicated with tacrolimus. Eur J Clin Pharmacol. 2012;68(12):1587-94. 7. Kasiske BL, Snyder JJ, Gilbertson D, Matas AJ. Diabetes mellitus after kidney transplantation in the United States. Am J Transplant. 2003;3(2):178-85. 8. Van Hooff JP, Christiaans MH, van Duijnhoven EM. Tacrolimus and posttransplant diabetes mellitus in renal transplantation. Transplantation. 2005;79(11):1465-9. Arch Endocrinol Metab. 2018;62/6


Diabetes after kidney transplantation

9. Numakura K, Satoh S, Tsuchiya N, Horikawa Y, Inoue T, Kakinuma H, et al. Clinical and genetic risk factors for posttransplant diabetes mellitus in adult renal transplant recipients treated with tacrolimus. Transplantation. 2005;80(10):1419-24. 10. Kim JS, Kim SK, Park JY, Kim YG, Moon JY, Lee SH, et al. Significant association between toll-like receptor gene polymorphisms and posttransplantation diabetes mellitus. Nephron. 2016;133(4): 279-86. 11. Bamoulid J, Courivaud C, Deschamps M, Mercier P, Ferrand C, Penfornis A, et al. IL-6 promoter polymorphism-174 is associated with new-onset diabetes after transplantation. J Am Soc Nephrol. 2006;17(8):2333-40. 12. Kim YG, Ihm CG, Lee TW, Lee SH, Jeong KH, Moon JY, et al. Association of genetic polymorphisms of interleukins with newonset diabetes after transplantation in renal transplantation. Transplantation. 2012 May 15;93(9):900-7. 13. Kang ES, Kim MS, Kim YS, Kim CH, Han SJ, Chun SW, et al. A polymorphism in the zinc transporter gene SLC30A8 confers resistance against posttransplantation diabetes mellitus in renal allograft recipients. Diabetes. 2008;57(4):1043-7. 14. Jeong K, Moon J, Chung J, Kim YH, Lee TW. Significant associations between CCL5 gene polymorphisms and posttransplantational diabetes mellitus in Korean renal allograft recipients. Am J Nephrol. 2010;32(4):356-61. 15. Ong S, Kang SW, Kim YH, Kim TH, Jeong KH, Kim SK, et al. Matrix metalloproteinase gene polymorphisms and new-onset diabetes after kidney transplantation in Korean renal transplant subjects. Transplant Proc. 2016;48(3):858-63. 16. Aktas A. Transplanted kidney function evaluation. Semin Nucl Med. 2014;44(2):129-45.

19. Shah T, Kasravi A, Huang E, Hayashi R, Young B, Cho YW, et al. Risk factors for development of new-onset diabetes mellitus after kidney transplantation. Transplantation. 2006;82(12):1673-6. 20. Kishi Y, Sugawara Y, Tamura S, Kaneko J, Matsui Y, Makuuchi M. New-onset diabetes mellitus after living donor liver transplantation: possible association with hepatitis C. Transplant Proc. 2006;38(9):2989-92. 21. Tueche SG. Diabetes mellitus after liver transplant new etiologic clues and cornerstones for understanding. Transplant Proc. 2003;35(4):1466-8. 22. Guitard J, Rostaing L, Kamar N. New-onset diabetes and nephropathy after renal transplantation. Contrib Nephrol. 2011;170:247-55. 23. Shrestha BM. Two Decades of Tacrolimus in Renal Transplant: Basic Science and Clinical Evidences. Exp Clin Transplant. 2017;15(1):1-9. 24. Muduma G, Saunders R, Odeyemi I, Pollock RF. Systematic review and meta-analysis of tacrolimus versus ciclosporin as primary immunosuppression after liver transplant. PLoS One. 2016;11(11):e0160421. 25. Sinangil A, Celik V, Barlas S, Sakaci T, Koc Y, Basturk T, et al. NewOnset Diabetes After Kidney Transplantation and Pretransplant Hypomagnesemia. Prog Transplant. 2016;26(1):55-61. 26. Miles AM, Sumrani N, Horowitz R, Homel P, Maursky V, Markell MS, et al. Diabetes mellitus after renal transplantation: as deleterious as non- transplant- associated diabetes? Transplantation. 1998;65(3):380-4. 27. Cosio FG, Kudva Y, van der Velde M, Larson TS, Textor SC, Griffin MD, et al. New onset hyperglycemia and diabetes are associated with increased cardiovascular risk after kidney transplantation. Kidney Int. 2005;67(6):2415-21.

CopyrightŠ AE&M all rights reserved.

17. D’Addio F, Vergani A, Di Fenza R, Tezza S, Bassi R, Fiorina P. Novel immunological aspects of pediatric kidney transplantation. G Ital Nefrol. 2012;29(1):44-8.

18. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2007;30 (suppl 1):s42-7.

Arch Endocrinol Metab. 2018;62/6

601


original article

Higher prevalence of permanent congenital hypothyroidism in the Southwest of Iran mostly caused by dyshormonogenesis: a five-year follow-up study Majid Aminzadeh1

ABSTRACT Division of Pediatric Endocrinology and Metabolism, Pediatric Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran 1

Correspondence to: Majid Aminzadeh Abuzar Children’s Hospital, Pasdaran Blvd, Ahvaz, Iran aminzadeh_m@ajums.ac.ir Received on Nov/12/2017 Accepted on May/30/2018 DOI: 10.20945/2359-3997000000085

Objective: The incidence of congenital hypothyroidism (CH) varies globally. This 5-year study aimed to determine the prevalence of permanent CH in the southwest of Iran. Materials and methods: Between January 2007 and December 2009, all newborns in Ahvaz, the biggest city in the southwest of Iran, were screened for CH using a heel-prick sample for thyrotropin (TSH) levels. Subjects with TSH ≥ 5 mU/L were evaluated for T4-TSH. Infants with T4 < 6.5 µg/dL, TSH > 10 mU/L, and normal T4 but persistent (> 60 days) high TSH were considered to have CH. After the third birthday, treatment was discontinued, and T4-TSH was reevaluated; subjects with TSH ≥ 10 mU/L were investigated using thyroid Tc99 scintigraphy (TS). Based on TS, they were classified as normal, dysgenetic, or athyretic (agenesis). Results: Screening was performed for 86,567 neonates, and 194 were confirmed to have CH (100 males; F/M = 0.94; overall incidence 1:446). After the third birthday, reevaluation was performed in all (except 18 that were not accessible). From 176 patients, 81 (46%) were diagnosed with permanent CH, and 95 were discharged as transient. Considering the same percentage in the lost cases, the prevalence of permanent CH was found to be 1:970. TS performed for 53 of the permanent subjects found agenesis/dysgenesis in 25 (F:M = 15:10) and a normal result in 28 (F:M = 11:17), indicating dyshormonogenesis as the cause in more than 50% of subjects. Conclusions: The incidence of CH in this area was found to be higher than that in other countries but less than the incidence rate reported in central Iran. The large number of transient cases of CH suggests environmental or maternal causes for the incidence rather than a genetic basis. Arch Endocrinol Metab. 2018;62(6):602-8 Keywords Congenital hypothyroidism; dyshormonogenesis; screening; Tc-99m thyroid scintigraphy; thyroid dysgenesis

INTRODUCTION

Copyright© AE&M all rights reserved.

S

creening for congenital hypothyroidism (CH), one of the most common causes of preventable mental retardation, has been performed in developing countries since the 1960s, but it has been routinely conducted in Iran only since the year 2000. The prevalence of CH has been reported to be 1/1,000 to 1/370 in Iran (1,2), which is significantly higher than that of the American and European countries (1/4,000) (3,4). The positive cost–benefit ratio of screening for CH ranges from 3.6:1 in developed countries (5) to 15:1 in developing countries such as Iran (6). Theoretically, dysgenesis, dyshormonogenesis, and iodine deficiency are the most common causes of CH. Prenatal (iodinated disinfectants) and postnatal iodine exposure (milk 602

iodine content) have also been implicated as etiologic factors (7,8). Varying incidence of CH has been reported in different areas of Iran, with no data from the southwest regions of the country, which is different from central and northern Iran in terms of weather, altitude, and gene pool. Another difference between various areas of Iran is that the southwest regions of the country have a higher rate of consanguineous marriages than the other regions. The latest reported CH incidence of 1/370 neonates in central Iran with a large number of transients (40%) (9) was significantly higher than the incidence rate previously reported. Accordingly, we aimed to determine the prevalence of CH in the southwest of Iran using a more definite diagnostic algorithm in a large enough population and Arch Endocrinol Metab. 2018;62/6


for a longer duration. The classic approach used herein may provide guidelines for diagnosing recalled (or referred) infants by a standard and safe algorithm (fully described in Materials and Methods and Table 1). Table 1. Study protocol for decision-making in the screening program of CH T4 (g/dL)

TSHa (mU/L)

Follow-up

≥ 8.5

< 5.5

Discharge

None

≤ 6.49

≥ 10

Treatment

As a CHb case

6.5–9.9

≥ 10

Suspected CH

TFT 2 wks later

6.5–9.9

5–9.9

Suspected CH

TFT 4 wks later

≥5

Suspected CH

TFT 4 wks later

> 6.0 after 60th day (10)

Treatment

As a CH case

≥ 10 Each level a

Decision

Thyroid stimulating hormone (normal for 2-20 wks = 0.5–5.5); b congenital hypothyroidism.

MATERIALS AND METHODS During the 3-year period from January 2007 to December 2009, all infants born in Ahvaz, the biggest city in the southwest of Iran, were enrolled (Table 2). This study was approved by the Ethical Committee of Ahvaz Jundishapur University of Medical Sciences. All neonates were screened before getting their identification card to ensure 100% compliance (however, some infants who did not pursue this policy were missed; Figure 1). Individuals who were born in Ahvaz but whose parents lived in other cities were excluded but were followed up on and treated if required. In all healthy and term babies, blood was obtained from a heel prick between the 3rd and 5th day of life and was collected on a filter paper for thyroid stimulating hormone (TSH) assessment using a kit (Padtan Elm Incorporation, Tehran, Iran) and an ELISA reader (Stat Fax 2100; Awareness Technologies, USA) by the same four staff members during the study period. For preterm infants and those who required postnatal hospitalization, testing was done after discharge but before the 30th day of life. The subjects were notified before the 10th day if the TSH level was > 5 mU/L and were referred for suspected CH. Screening in this study was based Table 2. Distribution of all screened newborns and CH patients, based on sex and year of birth Year of birtha

1st

2nd

All newborns

22 209 (51.2%)b

30 021 (50.5%)

34 337 (52%)

39 (48.7%)

85 (48.2%)

70 (55.7%)

CH subjects a

3rd

During the study period; numbers in parenthesis show the percentage of males. b

Arch Endocrinol Metab. 2018;62/6

on TSH measurement, so it may not have detected infants with delayed TSH elevation or with central or hypopituitary hypothyroidism (Figure 2). Infants with TSH > 5 mU/L but < 9.99 mU/L were retested by a second heel prick and referred if the level remained > 5 mU/L. Referred individuals were subjected to thyroid function tests (TFTs), including T4 and TSH, using a venous blood sample. The time line for performing the confirmatory TFT was chosen according to the TSH level observed in the first screening heel-prick test: a. For TSH < 20 mU/L, around the 21st day. b. For TSH between 20 and 39.9 mU/L, around the 14th day. c. For TSH ≥ 40 mU/L, reassessed and treated on the same day of referral. In the next years of screening, based on our national guidelines, the cutoff for emergency treatment was lowered to ≥ 20 mU/L. The confirmatory test was performed by venipuncture at least 2 hours after the last feeding in the morning. We used an ELISA kit (Monobind, USA) and an ELISA reader (BioTek ELx800, USA) to confirm or rule out CH among the referred suspected neonates. Dealing with the final laboratory results was done according to the protocol of the screening department of the Ministry of Health. This protocol was used with a more definite and detailed algorithm matching international standards so as to avoid unnecessary treatment of transient hyperthyrotropinemia (< 2 months) and to make treatment decisions for infrequent situations (Table 1). All referred suspected infants were followed up on until the final diagnosis was made: the beginning of treatment for CH or until the normalization of thyroid function in transient hyperthyrotropinemia. The aim was to begin replacement therapy with levothyroxine before the 30th day of life. The CH subjects younger than 6 months were followed up on monthly, between the 6th and 12th month, bimonthly, and after that, until the 36th month, quarterly. The goal was to maintain T4 at > 8 µg/dL, and TSH between 0.5 and 2.0 mU/L, based on the mean normal range (10-12), with normal growth and development. The treatment was stopped earlier than 3 years only if the subjects required < 12.5 µg/day of levothyroxine (1/4 of a 100 µg tablet, every other day) to stay in a euthyroid state. These subjects were classified as transient CH. Subclinical hypothyroidism was defined in the subjects who had normal T4 levels but persistent high TSH (5.5–15 mU/L) after the 60th day (13,14). The patients 603

Copyright© AE&M all rights reserved.

Congenital hypothyroidism in southwest of Iran


Congenital hypothyroidism in southwest of Iran

in whom treatment could be discontinued before 6 months (n = 2) were also classified as transient. However, classification of this group of subjects as transient high TSH would be more appropriate. To compare the final outcome based on the primary diagnostic TSH levels, all cases were classified into three groups: 1) ≥ 40 mU/L; 2) 20-39.9 mU/L; and 3) 5-19.9 mU/L (Table 3). After the third birthday, replacement therapy was discontinued for 1 month, then T4 and TSH assessments were performed in all subjects as reevaluation. Any rise of TSH to ≥ 10 mU/L in each test after discontinuation (also in those labeled as transient aged ≤ 3 years) was classified as permanent disease and was followed by thyroid scintigraphy (TS) and re-initiation of levothyroxine therapy. If the T4 and TSH concentrations were in the reference range, euthyroidism was assumed, and a

diagnosis of transient hypothyroidism was recorded. On the basis of the TS results, thyroid disorders were classified as agenesis (absence of thyroid on scan), dysgenetic (ectopia, thyroglossal cyst, lingual, hypoplasia, hemiagenesis, small thyroid, etc.), or dyshormonogenesis (normal scan or diffuse goiter; Figure 3). All those who could leave the treatment before 3 years of age or after the third year (after reevaluation) were followed up on for at least 24 months to distinguish between those with actual transient CH versus permanent subclinical hypothyroidism (TSH ≥ 10 mU/L). Data were presented after 5-year follow-up of all permanent and transient subjects (total 8-year duration of the study) to find the exact prevalence. Because most TSH results were reported in approximation (> 40 or > 100), the TSH ranges, rather than means, were used to compare the outcomes.

Figure 1. Three cases of congenital primary hypothyroidism were missed because of parental negligence, referred at 3 months with developmental delay.

Figure 2. A 6-month-old infant referred with poor growth diagnosed as congenital central hypothyroidism. He was missed because of screening with TSH. Serial photos (left to right: at the time of diagnosis, 1 week, 4 weeks, and 6 months after treatment) clearly show the facial changes as a reliable marker of clinical response over time.

Copyright© AE&M all rights reserved.

Table 3. Final outcome in 194 cases of CH according to their first serum TSH TSH*

Permanent; n (%)

F:M

Transient; n (%)

F:M

Unknown; n (%)

F:M

Total; n (%)

F:M

≥ 40

56 (52)†

30:26

39 (36)

19:20

13 (12)

6:7

108 (100)

55:53

20-39.9

10 (25.5)

4:6

25 (64)

11:14

4 (10.5)

2:2

39 (100)

17:22

< 20

15 (32)

6:9

31 (66)

15:16

1 (2)

1:0

47 (100)

22:25

Total

81 (42)

40:41

95 (49)

45:50

18 (9)

9:9

194 (100)

94:100

*mU/L; † percent of outcome in each TSH group.

604

Arch Endocrinol Metab. 2018;62/6


Figure 3. A neonate with goiterous congenital primary hypothyroidism (negative history of maternal thyroid disorder or perinatal risk factor) indicating dyshormonogenesis. She came back after 2 years because of developmental delay. She had a TSH > 100 mU/L due to not taking medication.

Statistics Data were analyzed using SPSS version 16.0 (SPSS Inc., Chicago, IL, USA), and the logistic regression test was used to analyze risk prediction in each outcome. P values < 0.05 were considered statistically significant.

RESULTS During the 3 years of screening, 86,567 neonates (51.28% male) were screened, of whom 5,923 (6.8%) had TSH levels ≥ 5 mU/L. Those with TSH ≥ 10 mU/L were referred directly, but a majority of them (n = 5,039; 85%), who had TSH values between 5 and 9.99 mU/L, were reassessed by the heel-prick method and then referred if the second test also revealed TSH to be > 5 mU/L (final recall rate referred on suspicion of CH = 2.4%; this means that up to 10 normal infants were recalled for testing for every 1 case of hypothyroidism). According to the acceptable definitions (Table 1), 194 cases (51.5% male) were diagnosed as CH (both permanent and transient), requiring treatment. The overall incidence was 1:446 live births with a female/male (F/M) ratio of 0.94. Table 2 shows the distribution by year of all live births and the prevalence of CH during the study period. After the third birthday, all 176 diagnosed CH subjects underwent reevaluation (18 of 194 were missed because of two deaths, five immigrations, and loss of contact information for the rest) with 4 weeks levothyroxine discontinuation and serum T4 and TSH measurement. Based on the TSH (if it was > 10 mU/L in the first or follow-up tests), 81 (46% of 176) subjects were diagnosed with permanent CH Arch Endocrinol Metab. 2018;62/6

(F:M = 40:41; 0.97, similar to the ratio seen in all 194 CH cases, including both permanent and transient). The remaining 95 subjects were followed up on with TSH rechecks for at least 2 more years with intervals of 3, 6, and 12 (and 24 if needed) months to make sure that there was no rise of TSH. These patients were discharged as transient CH. Considering the similar percentage of permanents in the missed group (46%), we probably lost at least 8 (of 18) more permanent CH cases to follow. The final prevalence of permanent CH was calculated as 1:970. The final outcomes were reviewed and compared in all cases based on their first diagnostic TSH values (Table 3). These data disclosed that 17 of 18 missed cases had TSH > 20 mU/L. This means that we probably lost more than 8 permanent CH cases among the 18 missed cases, indicating a higher prevalence of CH. Compared to those who had TSH < 20 mU/L (subclinical CH), the patients with TSH ≥ 40 mU/L had at least three times more risk to be permanent (1.41-6.22; 95% CI; OR). For those with TSH = 2039.9 mU/L, no significant risk was found (0.317-2.15; 95% CI; OR). TS performed in 53 of 81 permanents showed: agenesis in 5 (F:M = 2:3), dysgenesis in 20 (F:M = 13:7), and normal pattern in 28 (F:M = 11:17), indicating that > 50% of cases were caused by dyshormonogenesis. Details of the TS results are shown in Table 4. Compared to those who had TSH < 20 mU/L (subclinical CH), the patients with TSH ≥ 40 mU/L had almost five times more risk of having agenesis or dysgenesis in TS (0.98-28.2; 95% CI; OR). For those with TSH = 20-39.9 mU/L, no significant risk was found (0.2-19.9; 95% CI; OR). There were 11 subjects receiving treatment with the diagnosis of mild subclinical CH (TSH = 5.5-10). On reevaluation, four cases were found to be permanent: two boys (both normal scan) and two girls (one dysgenesis and one normal scan). The 7 other subjects were discharged as transient. During this period, two cases of pan-hypopituitarism (normal ACTH, adrenocorticotropic hormone), two with genetically confirmed Pit-1 defect (deficiency of TSH, growth hormone, and prolactin), and one with isolated TSH deficiency (Figure 2) were also diagnosed independent of the screening program and were included in the overall prevalence without performing TS. 605

Copyright© AE&M all rights reserved.

Congenital hypothyroidism in southwest of Iran


Congenital hypothyroidism in southwest of Iran

Table 4. Comparison of the first serum TSH values in different etiologies of permanent CH based on TS TS group

TSH ≥ 40*

Agenesis Dysgenesis Normal Total

F:M

20-39.9

F:M

TSH < 20

F:M

4 (80)†

2:2

1 (20)

17 (85)

11:6

1 (5)

16 (57)

8:8

37 (69.8)

21:16

Total

F:M

0:1

0 (0)

0:0

5 (100)

2:3

1:0

2 (10)

1:1

20 (100)

13:7

4 (14.3)

1:3

8 (25)

2:6

28 (100)

11:17

6 (11.3)

2:4

10 (18.8)

3:7

53 (100)

27:26

*mU/L; † percent of TSH group in each outcome.

Copyright© AE&M all rights reserved.

DISCUSSION In concurrence with previous reports, we observed a high prevalence of CH in our study, similar to other Asian populations. As per the Iranian Ministry of Health recommendation and our study design, we used TSH as the screening assay to evaluate CH. This probably resulted in missing only 2 of 100 CH cases (12), including those with secondary or tertiary hypothyroidism, infants with an absence of the free T4 feedback mechanism, and those with a delayed elevation of TSH levels. As mentioned above, we found 5 cases of central CH that their missing was unavoidable (Figure 2). Accordingly, the ideal screening method would be to evaluate primary TSH with T4, but it is cost-prohibitive. As with most congenital disorders, the main causes of the differences in CH prevalence would be ethnicity, gene pool, and the rate of consanguineous marriages (15,16). Similar to other Asian populations, our neonates are at a higher risk for CH, although the main etiologic factors including dysgenesis or dyshormonogenesis were not very prevalent in our cases of CH. In fact, similar to other Iranian studies (17), we found a high rate of transient CH cases (near half), which cannot be attributed to genetics but rather to maternal iodine supplementation and/ or thyroid status (7,8). While early treatment of even transient subjects is mandatory to prevent any degree of neurodevelopment impairment, better management of iodine supplementation and maternal thyroid status can obviate a large number of recalls, unnecessary venipuncture, replacement therapy, clinic visits, and, more importantly, family stress and conflict. After a 2-year follow-up of all those who could leave treatment before or after 3 years of age (including those with mild subclinical CH; TSH < 10 mU/L), we found a higher rate of transients than that previously reported in Isfahan (9) (≈ 50% vs. 40%). Tc-99m TS was reported to be a useful diagnostic tool for the investigation of suspected CH and can 606

potentially help manage and predict the lifelong replacement therapy requirement (18). However, in the first 3 years of life, the treatment plan remains the same. The prevalence of permanent CH was found to be about 1/970 live births, which is less than the 1/750 live births reported in Isfahan (9) but near to the 1/918 live births identified in Asian families living in England (19). Using a standard classic approach with a huge number of live births, the prevalence rate of 1/446 (total of permanent and transient cases) found in our study supports the controversial report from central Iran showing a CH incidence of 1/370 (2). The female-to-male ratio for all CH subjects in our study (0.94) was quite similar to that of the whole country (20). Additionally, many cases of permanent subclinical CH with seemingly normal TS results (except one with a right hemiagenesis) were identified in our study. Because the benefits of replacement therapy are significant even in adults (21), it is logical to treat and follow up on all subclinical CH cases, particularly those with TSH ≥ 10 mU/L. Although patients with TSH levels > 10 mU/L often have reduced free T4 levels and may have hypothyroid symptoms, theoretically, they must be clinically normal regarding the normal T4 levels. The high incidence of permanent subclinical CH in cases with normal T4 but mildly elevated TSH (5–10 mU/L) shows the importance of treatment and followup for such infants. Interestingly, only permanent CH subjects with agenesis/dysgenesis in their scan had a female predominance, similar to what was discovered in CH subjects of Western countries (mostly caused by dysgenesis). The finding of a F:M ≈ 1 in all subjects (permanent and transient cases) and a male predominance in the dyshormonogenesis group is compatible with their etiologic background. Transient CH subjects as a big part of total CH cases possibly caused by environmental factors should not have a Arch Endocrinol Metab. 2018;62/6


Congenital hypothyroidism in southwest of Iran

Acknowledgments: this study was performed as a research project (ID: U-87010) approved by the vice chancellor of the Research Center at Ahvaz Jundishapur University of Medical Sciences. The author would like to thank all the staff of the screening department in the East and West Health Centers of Ahvaz. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Ordookhani A, Mirmiran P, Moharamzadeh M, Hedayati M, Azizi F. A high prevalence of consanguineous and severe congenital hypothyroidism in an Iranian population. J Pediatr Endocrinol Metab. 2004;17(9):1201-9. Arch Endocrinol Metab. 2018;62/6

2. Hashemipour M, Amini M, Iranpour R, Sadri GH, Javaheri N, Haghighi S, et al. Prevalence of congenital hypothyroidism in Isfahan, Iran: results of a survey on 20,000 neonates. Horm Res. 2004;62(2):79-83. 3. Deladoey J, Belanger N, Vliet GV. Random variability in congenital hypothyroidism from thyroid dysgenesis over 16 years in Québec. J Clin Endocrinol Metab. 2007;92(8):3158-61. 4. Rastogi MV, LaFranchi SH. Congenital hypothyroidism. Orphanet J Rare Dis. 2010;5:17. 5. Gu X, Wang J,Ye J, Cheng X. [A cost-benefit evaluation of neonatal screening for phenylketonuria and congenital hypothyroidism]. Zhonghua Yu Fang Yi Xue Za Zhi. 2000;34(3):147-9. 6. Delavari AR, Ahmadi SY, Birjandi R, Mahdavi AR, Nejad AN, Dini M. Cost-benefit analysis of the neonatal screening program implementation for congenital hypothyroidism in I. R. Iran. Int J Endocrinol Metabol. 2006;4(2):84-7. 7. Ordookhani A, Pourafkari M, Mirmiran P, Neshandar-Asl I, Fotouhi F, Hedayati SM, et al. Etiologies of transient congenital hypothyroidism in Tehran and Damavand. Iran J Endocrinol Metabol. 2004;6(2):107-13. 8. Nasri P, Hashemipour M, Hovsepian S, Amini M, Heidari K, Sajjadi SA, et al. Comparison of urine and milk iodine concentration among congenitally hypothyroid neonates and their mothers and a control group. Iran J Endocrinol Metabol. 2009;11(3):265-72. 9. Hashemipour M, Hovsepian S, Kelishadi R, Iranpour R, Hadian R, Haghighi S, et al. Permanent and transient congenital hypothyroidism in Isfahan-Iran. J Med Screen. 2009;16(1):11-6. 10. Lem AJ, de Rijke YB, van Toor H, de Ridder MA, Visser TJ, HokkenKoelega AC. Serum thyroid hormone levels in healthy children from birth to adulthood and in short children born small for gestational age. J Clin Endocrinol Metab. 2012;97(9):3170-8. 11. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab. 2002;87(2):489-99. 12. American Academy of Pediatrics, Rose SR; Section on Endocrinology and Committee on Genetics, American Thyroid Association, Brown RS; Public Health Committee, Lawson Wilkins Pediatric Endocrine Society, Foley T, Kaplowitz PB, Kaye CI, Sundararajan S, Varma SK. Update of newborn screening and therapy for congenital hypothyroidism. Pediatrics. 2006;117(6):2290-303. 13. Gillet M. Subclinical hypothyroidism: subclinical thyroid disease: scientific review and guidelines for diagnosis and management. Clin Biochem Rev. 2004;29(3):191-4. 14. Surks MI, Ortiz E, Daniels GH, Sawin CT, Col NF, Cobin RH, et al. Subclinical thyroid disease: scientific review and guidelines for diagnosis and management. JAMA. 2004;291(2):228-38. 15. Hall SK, Hutchesson AC, Kirk JM. Congenital hypothyroidism, seasonality and consanguinity in the West Midlands, England. Acta Paediatr. 1999;88(2):212-5. 16. Hashemipour M, Amini M, Talaie M, Kelishadi R, Hovsepian S, Iranpour R, et al. Parental consanguinity among parents of neonates with congenital hypothyroidism in Isfahan. East Mediterr Health J. 2007;13(3):567-74. 17. Karamizadeh Z, Dalili S, Sanei-far H, Karamifard H, Mohammadi H, Amirhakimi G. Does congenital hypothyroidism have different etiologies in Iran? Iran J Pediatr. 2011;21(2):188-92. 18. Iranpour R, Hashemipour M, Amini M, Talaei SM, Kelishadi R, Hovsepian S, et al. [Tc]-99m thyroid scintigraphy in congenital hypothyroidism screening program. J Trop Pediatr. 2006;52(6):411-5. 19. Rosenthal M, Addison GM, Price DA. Congenital hypothyroidism: increased incidence in Asian families. Arch Dis Child. 1988;63(7):790-3.

607

Copyright© AE&M all rights reserved.

sex difference. Dyshormonogenesis is almost always inherited in a recessive pattern, but only 2% of dysgenesis is familial (22). This may explain the F:M differences we discovered in two groups of thyroid dyshormonogenesis and thyroid dysgenesis. In concordance with previous studies (17,23), in our study, patients with permanent CH had higher TSH levels than transient ones during the neonatal period. This finding emphasizes the need for careful follow-up in those with TSH ≥ 40 mU/L. The limitations of this study include inexperienced laboratory technicians, health staff, and physicians, particularly in the first year of the study (24,25); noncooperative hospitals; families who did not participate or who approached private clinics independently; and the inability to achieve 100% screening coverage. Table 2 displays the number of screened newborns, rather than live births, which may have resulted in underestimating the total number of CH cases, although this problem theoretically does not change the prevalence recorded by our study. The strength of this study is that it determined the real percentage of transient CH cases and also identified those cases with permanent CH but normal TS with a high degree of accuracy. In conclusion, the prevalence of CH in the southwest part of Iran was found to be much higher than that of the Western countries; however, it was less than that found in central Iran and similar to that seen in Asian families living in Western countries. Accordingly, due to the importance of replacement therapy for transient cases, adequate follow-up is necessary. The high incidence of transient cases suggests environmental and/or maternal causes, rather than a genetic basis for the CH.


Congenital hypothyroidism in southwest of Iran

20. Veisani Y, Sayehmiri K, Rezaeian S, Delpisheh A. Congenital hypothyroidism screening program in iran; a systematic review and metaanalysis. Iran J Pediatr. 2014;24(6):665-72.

23. Ordooei M, RABIEi A, Soleimanizad R, Mirjalili F. Prevalence of permanent congenital hypothyroidism in children in Yazd, Central Iran. Iran J Public Health. 2013;42(9):1016-20.

21. Meier C, Staub JJ, Roth CB, Guglielmetti M, Kunz M, Miserez AR, et al. TSH-controlled L-thyroxine therapy reduces cholesterol levels and clinical symptoms in subclinical hypothyroidism: a double blind, placebo-controlled trial (Basel Thyroid Study). J Clin Endocrinol Metab. 2001;86(10):4860-6.

24. Korada M, Kibirige M, Turner S, Day J, Johnstone H, Cheetham T. The implementation of revised guidelines and the performance of a screening programme for congenital hypothyroidism. J Med Screen. 2008;15(1):5-8. 25. Jones JH, Mackenzie J, Croft GA, Beaton S, Young D, Donaldson MD. Improvement in screening performance and diagnosis of congenital hypothyroidism in Scotland 1979-2003. Arch Dis Child. 2006;91(8):680-5.

CopyrightŠ AE&M all rights reserved.

22. Agrawal P, Philip R, Saran S, Gutch M, Razi MS, Agroiya P, et al. Congenital hypothyroidism. Indian J Endocrinol Metab. 2015;19(2):221-7.

608

Arch Endocrinol Metab. 2018;62/6


original article

Conversion to Graves disease from Hashimoto thyroiditis: a study of 24 patients Beatriz Gonzalez-Aguilera1, Daniela Betea2, Laurence Lutteri3, Etienne Cavalier3, Vincent Geenen2, Albert Beckers2, Hernan Valdes-Socin2

ABSTRACT Objective: The conversion of Hashimoto’s thyroiditis (HT) to hyperthyroidism due to thyrotropin receptor antibodies is intriguing and considered rare. The contribution of TSH receptor blocking antibodies (TRAb), which may be stimulators (TSAb) or blockers (TBAb), is suspected. We describe clinical and biological variables in a series of patients switching from Hashimoto’s thyroiditis to Grave’s disease. Subjects and methods: Retrospective case study of 24 patients with Hashimoto’s thyroiditis followed during 48 ± 36 months that developed later Graves’ disease (GD). These variables were analysed in the hypo and hyperthyroid phase: age, sex, initial TSH, free triiodothyronine (fT3), free thyroxine (fT4), anti-TPO, TBII antibodies, parietal cell autoantibodies, time between hypo and hyperthyroidism, thyroid volume and levothyroxine doses (LT). Results: In HT, mean TSH was 9.4 ± 26.1 UI/L and levothyroxine treatment was 66.2 ± 30.8 µg/day. The switch to GD was observed 38 ± 45 months after HT diagnosis. As expected, we found significant differences on TSH, FT3, FT4 and TBAb levels. Three out of 14 patients had parietal cell autoantibodies. In two of these three cases there was an Helicobacter pylori infection. There were no significant differences between HT and GD groups with respect to thyroid volume. Conclusions: To our knowledge, large series documenting the conversion of HT to GD are scarce. Although rare, this phenomenon should not be misdiagnosed. Suspicion should be raised whenever thyroxine posology must be tapered down during the followup of HT patients. Further immunological and genetic studies are needed to explain this unusual autoimmune change. Arch Endocrinol Metab. 2018;62(6):609-14

Department of Endocrinology, University Hospital Juan Ramón Jiménez (Huelva), Quirónsalud Hospital Group, Sevilla, Spain 2 Department of Endocrinology, CHU, Liège, Belgium 3 Department of Clinical Biology, CHU, Liège, Belgium 1

Correspondence to: Hernan Valdes-Socin hg.valdessocin@chuliege.be Received on May/10/2018 Accepted on Oct/19/2018 DOI: 10.20945/2359-3997000000086:

INTRODUCTION

H

ashimoto’s thyroiditis (HT) and Graves’ disease (GD) are autoimmune thyroid disorders (AITDs) with different physiopathology, being traditionally regarded as two different disease entities. More recent views, in contrast, have considered the hypothesis that there might be a continuum between HT and GD (1,2). Recently, it has been sporadically reported that GD and HT may follow one another in the same individuals due to a sequential phenotypic conversion from GD to HT or vice versa (3,4). The most common scenario is the evolution from GD into HT, whereas the switch from HT into GD seems to be less common. This is probably due to the lack of a critical mass of functioning thyroid tissue able to react to thyrotropin (TSH) receptor autoantibodies (TRABs) in individuals with long-standing HT (5). The phenomenon of the switch from Hashimoto’s thyroiditis to hyperthyroidism and/or Graves’ Arch Endocrinol Metab. 2018;62/6

ophthalmopathy is also known as ‘hypothyroid Graves’ disease’ being first described by Wyse and cols. in 1968 (6). Since the first description of these cases was given, the concept of hyperthyroidism as an essential component of Graves’ disease has been modified. For 50 years, several similar cases have been published. However, texts and medical books hardly refer to this phenomenon. Moreover, the pathogenic mechanism is not well known (6-11). One of the most accepted hypotheses is the potential contribution of blocking antibodies (TBAb) and TSH receptor stimulators (TSAb) (8-13). TSAb antibodies are responsible for Graves’ disease hyperthyroidism while TBAb antibodies are sometimes responsible for a pattern of hypothyroidism. The change of antibodies from TBAb to TSAb (or vice versa) occurs rarely in patients treated with levothyroxine for hypothyroidism or in patients with Graves’ disease treated with antithyroid drugs. These changes involve differences in the concentrations, 609

Copyright© AE&M all rights reserved.

Keywords Hashimoto’s thyroiditis; TBII; Graves hyperthyroidism; TSH receptor antibodies; autoimmune conversion


Conversion to Graves disease from Hashimoto thyroiditis

Partial (weak) agonist Neutral Receptor constitutive activity

Hypothyroidism

TB Ab/Ts Ab activity

Full (strong) agonist

Hyperthyroidism

affinities and potency of TSAb and TBAb. Thus, the increase of TSAb after treatment with levothyroxine may be sufficient to counteract the TBAb antibodies and result in hyperthyroidism (Figure 1). In the present study, we describe the clinical, immunological and biochemical characteristics of patients with HT, who later developed GD. We discuss our results and compare them with the available literature.

Mean time from hypothyroidism to hyperthyroidism in this series: 38 months

Figure 1. The figure describes the transition from hypothyroidism to hyperthyroidism during follow up in the same patient. The phenomenon is due to a shift in the concentrations and affinities of TBAb and TSAb. In our series the mean time for this switch was of 38 months. Thyrotropin (TSH) has a constitutive activity, as shown by the horizontal line through the wedge. TSHR ligand (TSH or TSAb) further increase receptor activity. An inverse agonist supresses constitutive activity, whereas a full agonist maximally activates the receptor. Ligands of intermediate activity are either neutral agonists or partial agonists or inverse agonists. Ligands can also be an antagonist depending on their relative affinities and binding sites. Therefore, TSAb beeing a partial agonist may also be an antagonist for TSH. If a serum displays both TSAb and TBAb activity, unless the former is very weak and the latter is very strong, it cannot be assumed that there are two separate antibodies. [Adapted from: McLachlan et al. (13)].

MATERIALS AND METHODS

Copyright© AE&M all rights reserved.

Design This study is an observational retrospective case study performed in the Centre Hospitalier Universitaire de Liège. Among nearly 2000 patients with HT followed up in our endocrine unit during the period 20002016, we identified a group of 24 patients. These patients represented 1.2% of the series. Moreover, the 24 patients performed a posterior switch to GD. All the data recorded at HT diagnosis and at GD presentation were anonymously reconstructed after informed consent, from patients’ files.

Eligibility criteria The inclusion criteria were as follows: males and females with HT and age ≥ 18. The criteria for the diagnosis of HT 610

were: a) Elevated TSH levels > 4 mUI/L, the presence of serum thyroid peroxidase autoantibodies (TPOAbs) at titres above the upper limits of the reference ranges; b) hypoechogenic thyroid pattern at ultrasonography (US) consistent with AITD. The absence of ThyroidBinding Inhibitory Immunoglobulin (TBII) was determined in only three patients. The criteria for diagnosis of GD were as follows: a) elevated levels of free thyroxine (T4) and suppressed TSH; b) the presence of TBII; c) hypoechogenic thyroid pattern at US consistent with AITD; d) no tendency to spontaneous normalization of hyperthyroidism before methimazole treatment; and e) positive technecium99/ iodine123 thyroid scintigraphy. The exclusion criteria were as follows: hypothyroidism due to other causes, and patients < 18 years.

Assessment tools and variables The following variables were collected: age, sex, TSH, free T3 and free T4 at the beginning of the study and at the moment of the switch to hyperthyroidism, anti-TPO and TBII antibody titres in the hypo and hyperthyroid phase, time evolution of hypothyroidism, time between hypo and hyperthyroidism, levothyroxine doses (LT) in both groups, presence or absence of Graves’ ophthalmopathy, positivity for anti-parietal cell antibodies and type of treatment for hyperthyroidism.

Laboratory tests Serum concentrations of TSH (normal range 0,27– 4,2 mU/L), fT4 (normal range 11,6-21,9 pmol/L) and fT3 (normal range 3-6,8 pmol/L) were measured by electrochemiluminescence methods (using system Cobas e801, Roche®) (14-16). Anti-TPO antibodies were measured by electrochemiluminescence immunoassay methods, (using Cobas e 411, Cobas e 601 and Cobas e 602, Roche®) (17). According to the employed method, anti-TPOAbs values above 34 IU/ml are defined as positive. TBAb antibodies (Thyroïd Receptor Binding antibodies) (18) were evaluated at baseline and at the hyperthyroid phase by electrochemiluminescence methods, using the system Cobas e801, Roche®. This a quantitative third generation TBAb method, in which patient’s serum autoantibodies inhibit, in vitro, the fixation of a human monoclonal stimulating antibody (M22) against TSH Receptor (TSHR). According to the employed method, values above 1,2 IU/L are considered as positive. Arch Endocrinol Metab. 2018;62/6


Conversion to Graves disease from Hashimoto thyroiditis

Antiparietal cell antibodies research was based on indirect immunofluorescence methods (Menarini®). The screening dilution is 1/20. The results are reported as negative with a titer less than the screening dilution, positive with a titer greater than or equal to the screening dilution or, preferably, positive with specific endpoint titer. Thyroid Ultrasounds examinations for assessment of echogenicity were always performed by experienced ultrasonographers (DB, HVS) with high–resolution machines. With regard to thyroid function patterns at diagnosis of both HT and GD, they were evaluated according to fT4 and TSH serum levels and classified into the following groups: 1) euthyroidism (both fT4 and TSH within normal limits); 2) hypothyroidism (low fT4, as opposed to elevated TSH) 3) overt hyperthyroidism (suppressed TSH, as opposed to elevated fT4) and 4) subclinical hyperthyroidism (supressed TSH, as opposed to normal fT4).

Statistical analysis Data were analysed using IBM SPSS statistics version 22 Software. The normality of the variables was estimated using the Shapiro test and Kolmogorov-Smirnov test. A descriptive statistics table of all variables was obtained, taking the median, range, mean and standard deviation among other parameters, as well as a table of frequencies.

RESULTS Twenty-four patients (22 women, 2 men) had hypothyroidism and HT followed by a phase of

hyperthyroidism. They were followed up during a mean time of 48 ± 36 months. During hypothyroidism, mean levothyroxine treatment was 66.2 ± 30.8 µg/day. Following HT diagnosis, patients developed GD, after a mean time of 38 ± 45 months. None of the HT patients had initially features of Graves ophtalmopathy. Levothyroxine was stopped in all patients when hyperthyroidism was suspected. Suspicion of hyperthyroidism was raised because of frequent reduction of levothyroxine or the development of overt biological hyperthyroidism during follow up in some cases. The baseline characteristics and comparative descriptive statistics are shown in Table 1. Patients developed hyperthyroidism with mean TRAb levels of 7.9 ± 7.1 IU/L. One hypothyroid patient developed hyperthyroidism three months postpartum and another hypothyroid patient was diagnosed with Graves disease during the first trimester of her four pregnancy. Hyperthyroidism was treated with radioiodine in 7 patients and with antithyroid drugs in 17 patients. Mean time to resolve hyperthyroidism in the radioiodine group was 6 ± 1 months whereas in the methimazole group it was 18 ± 2 months. After Graves disease, all patients were hypothyroid. They needed a mean levothyroxine treatment of 85 ± 20 µg/day. During the hyperthyroid phase follow-up, four patients developed moderate Graves’ ophthalmopathy and three patients (10%) had anti-parietal cell antibodies. These three patients had a moderate gastritis pattern at gastric biopsy, without gastric atrophy. Helicobacter pylori were found in two of the three

Table 1. Descriptive statistics of 24 cases of patients that developed Graves disease after Hashimoto Thyroiditis Hashimoto Thyroiditis hypothyroidism

Graves hyperthyroidism

SD

Median

Range

Mean

SD

Median

Range

p

Age at diagnosis (years)

39

13

39

18

42

11.3

42

45

ns

Levothyroxine dose (µg/day)

66

30

58.7

125

na

na

na

na

na

Thyroid volume (mL)

7.4

3.1

7.2

12.3

7.38

3.1

5.2

12

ns

TSH (mUI/L)

9.4

26.1

3

12.2

< 0.01

-

< 0.01

-

< 0.05

FT3 (pmol/L)

3.6

1.8

4.7

12

7.9

8.8

24

< 0.05

FT4 (pmol/L)

8.7

5.1

10.5

13.5

34

32

24.5

31

< 0.05

Anti TPO antibodies (IU/mL)

383

988

146

4735

335

240

273

580

ns

TBII (IU/L)

nd

nd

nd

nd

7.9

7.1

4.8

13

na

Switch from HT to GD (months)

na

na

na

na

38

45

18

168

na

Copyright© AE&M all rights reserved.

Mean

nd: not done; ns: non significant; na: not applicable. Arch Endocrinol Metab. 2018;62/6

611


Conversion to Graves disease from Hashimoto thyroiditis

Table 2. Conversion from Hashimoto to Graves’ disease: reported series of adult patients Number of patients

Mean age

Time to develop hyperthyroidism from hypothyroidism

Osorio-Salazar (19)a

7

33

Takasu and cols.b (21)

2

45

Takeda and cols. (23)

1

Cho and cols. (24)

TSAb Positive (n cases)

TBAb Positive (n cases)

Mean Anti TPO (IU/mL)

36 months

Positive (hyper)

Negative (Hyper)

414

NA

110 (Hypo) 900 (Hyper)

98 (Hypo) 20 (Hyper)

NA

48

36 months

93 (Hypo) 163 (Hyper)

96 (Hypo) 31 (Hyper)

105

1

40

12 months

92 (Hypo) 2703 (Hyper)

89 (Hypo) 12 (Hyper)

6400

Takasu and cols. (25)

8

38

NA

92 (Hypo) 1490 (Hyper)

96 (Hypo) 2 (Hyper)

800

Kraiem and cols. (26)

1

55

36 months

576 (Hyper)

Negative (Hyper)

NA

This series

24

36.1

38 months

Not done

24

228

All patients were treated with LT4. Values for all thyroid autoantibodies are expressed in μU/mL; Neg, negative (undetectable); NA; not available. a In this study TSAb and TBAb values were only registrated in two cases out of seven patients. b Only two patients of 34 described by Takasu and cols. (21) had hypothyroidism due to TBAb and a switch to TSAb during hyperthyroidism. Hyper: hyperthyroidism; hypo: hypothyroidism; TBAb: TSH blocking autoantibodies; TPOAb: autoantibodies to thyroid peroxidase; TSAb: thyroid-stimulating autoantibodies.

patients. As expected, levels of fT3, fT4 and TRAb were significantly higher and TSH levels was significantly lower at the hyperthyroid state (p < 0.05). There were no significant differences regarding the mean thyroid volume in hypothyroid nor in hyperthyroidism state.

Copyright© AE&M all rights reserved.

DISCUSSION The conversion of autoimmune hypothyroidism to Graves’ hyperthyroidism is a rare clinical situation that is seldom described in text books and cases reports in the literature (1,13,19). This is one of the largest adult series of reported cases in which the conversion of hypothyroidism to hyperthyroidism was well documented (Table 2). The switch from HT to GD depends on two TSH receptor binding antibodies with different effect in thyroid cells. Firstly, TSH receptor stimulating antibodies (TSAb) are responsible for Graves’ disease. Secondly, TSH receptor antibodies with inhibitory activity (TBAb) cause, unfrequently, a situation of hypothyroidism. The mechanism of TBAb hypothyroidism is different from the one caused by destructive Hashimoto’s thyroiditis (8). The nomenclature for the methods used in the measurement of the antibodies against the TSH receptor is confusing. In short, TBII (TSAb and TBAb) antibodies measure the response of antibodies to TSHR, but they do not differentiate the functional activity (whether they are stimulatory, blocking or neutral). 612

Instead, TSI antibodies measure functionality, as they stimulate TSHR receptor through the production of cAMP (12,20). As we can see in Figure 1, depending on the affinity of the ligand, we can find in the same patient a predominance of the stimulatory or the blocking action of the receptor, thus resulting in hypo or hyperthyroidism (13). One limiting factor of our study is the retrospective design. Another limiting factor is that TBAb were not systematically studied, as no apparent reason was present to dose these antibodies in hypothyroid patients. However, Takasu and cols studied TBAb and TSAb in 34 TBAb positive known patients with hypothyroidism. They demonstrated during follow up that with the disappearance of TBAb, there was recovery from hypothyroidism in 13 (87%) of these patients. Moreover, two of the 34 (5.9%) TBAb-positive patients with hypothyroidism developed TSAb-positive Graves’ hyperthyroidism (21). Osorio-Salazar and cols reported in 1994 a French series of seven females with HT who presented after a few months or years signs of Graves’ disease. Retrospectively, TSAb were found in some of the patients that switched form hypothyroidism into hyperthyroidism (19). There are several mechanisms involved in the switching from TBAb to TSAb or vice versa. These include effects and circumstances that affect the autoimmune response, such as levothyroxine and antithyroid Arch Endocrinol Metab. 2018;62/6


Conversion to Graves disease from Hashimoto thyroiditis

Arch Endocrinol Metab. 2018;62/6

other have already reported (31-33). The interest of diagnosing autoimmune gastritis in these patients is to evaluate the possible impact of hypoclorydia in levothyroxine and micronutriments (iron, vitamin B12) malabsorption. In two of our patients, Helicobacter pilory was found in gastric biopsies, suggesting its role in gastric autoimmunity. Moreover, a possible effect of Helicobacter pilory infection has been suggested in thyroid autoimmunity because of cross autoantigens reaction between the bacteria capsid and gastric cells (31-33). In conclusion, conversion of Hashimoto Thyroiditis towards Graves’ disease is still an underestimated clinical feature for most clinicians. In our series, this phenomenon was observed in 1.2% of patients. Interestingly, the conversion from HT to GD has been described as prevalent (25.7% of cases) in a controlled series of 35 children with either Down or Turner Syndrome (33). Our data, as well as previous reported cases should raise the suspicious of this phenomenon whenever hyperthyroidism and difficulties in equilibrating patients are observed in “usual” autoimmune hypothyroid patients supplemented with levothyroxine. Several mechanisms have been postulated behind the conversion from HT to GD. One of them is the presence of different autoantibodies that include thyroid stimulating antibodies and thyroid blocking antibodies. Another possible explanation for this transition is that the autoimmune tissue damage, initially severe enough to cause thyroid hypofunction, recovers sufficiently to allow subsequent stimulation by TSAb. More extensive cohort immunological and genetic studies are necessary to gain insight into these interesting observations. Acknowledgements: we thank the Fonds Léon Frédéricq for its support. The Prize of Fondation Jaumain was attributed to Dr. Hernan Valdes-Socin for his clinical and research activities. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Wood LC, Ingbar SH. Hypothyroidism as a late sequela in patient with Graves’ disease treated with antithyroid agents. J Clin Invest. 1999;64:1429-36. 2. Sofie Bliddal, Claus Henrik Nielsen, Ulla Feldt-Rasmussen. Recent advances in understanding autoinmune thyroid disease: the tallest tree in the forest of polyautoimmunity. F1000 Research. 2017;7:1-12.

613

Copyright© AE&M all rights reserved.

drug replacement therapy, immunosuppression/ hemodilution occurring during pregnancy, and the disappearance of this postpartum immune tolerance and inherent properties of TSAb and TBAb (13,21). These authors studied whether there was a correlation between the dose of treatment with levothyroxine and the levels of anti-TPO antibodies with the development of TBII antibodies and subsequent hyperthyroidism. They found a significant association with anti-TPO antibodies levels, but no association with the dose of levothyroxine was found (13,21). The effect of LT treatment on thyroid autoimmunity is an old ‘workhorse’ with countless reports in the literature for more than 50 years (8). Recent longitudinal studies suggest that the effect of LT treatment has little or no effect on anti-TPO and anti-thyroglobulin antibodies in euthyroid patients or in patients with subclinical hypothyroidism. The effect of LT on hypothyroid patients is more debated. Takasu and Matshushita’s studies have reported a positive correlation between LT dose and the appearance of de novo TSAb autoantibodies or increased TSAb antibody levels (21). They also described the development of hyperthyroidism in hypothyroid patients. This association may simply be fortuitous or it can be explained by the regulatory effect of thyroid hormones on the innate and adaptive immune response: Dendritic cells modulate their phenotype under the influence of elevated levels of thyroid hormone (27). Another possible explanation for this transition from hypothyroidism to hyperthyroidism (21-28) is that the autoimmune tissue damage (initially severe enough to cause thyroid hypofunction), recovers sufficiently to allow subsequent stimulation by TSAb (28). This expansion of the autoimmune response has been called ‘determinant spreading’, where a large number of different T cell clones attack a broader range of determinants of the invading pathogens or the inflamed tissues, instead of focusing on few immunodominant determinants (29,30). In this sense, it seems that longstanding hypothyroidism leads to increased and permanent tissue damage and there is likely less functioning thyroid tissue able to react to thyrotropin receptor autoantibodies as compared to hypothyroidism of short evolution (5,29,30). Finally, we detected a thyroid and a gastric autoimmunity in three of 14 (21%) patients. Asymptomatic autoimmune gastritis is prevalent in patients with autoimmune thyroiditis as we and


Conversion to Graves disease from Hashimoto thyroiditis

3. Umar H, Muallima N, Adam JM, Sanusi H. Hashimoto’s thyroiditis following Graves’ disease. Acta Med Indones. 2010;42:31-5. 4. Champion B, Gopinath B, Ma G, El-Kaissi S, Wall JR. Conversion to Graves’ hyperthyroidism in a patient with hypothyroidism due to Hashimoto’s thyroiditis documented by real-time thyroid ultrasonography. Thyroid. 2008; 18:1135-7. 5. Kamath C, Young S, Kabelis K, Sanders J, Adlan MA, Furmaniak J, et al. Thyrotrophin receptor antibody characteristics in a woman with long-standing Hashimoto’s who developed Graves’ disease and pretibial myxoedema. Clin Endocrinol. 2012; 77:465-70. 6. Wyse EP, McConahey WM, Woolner LB, Scholz DA, Kearns TP. Ophthalmopathy without hyperthyroidism in patients with histologic Hashimoto’s thyroiditis. J Clin Endocrinol Metab. 1968;28(11):1623-9. 7. Bell PM, Sinnamon DG, Smyth PP, Drexhage HA, Haire M, Bottazzo GF, et al. Hyperthyroidism following primary hypothyroidism in association with polyendocrine autoimmunity. Acta Endocrinol (Copenh). 1985;108(4):491-7. 8. BH, Kang. Hyperthyroidism following hypothyroidism-a case report. Med J Malaysia. 1991;46(3):287-9. 9. Kasagi K, Konishi J, Iida Y, Mori T, Torizuka K. Changes in thyroid-stimulating and TSH-binding inhibitory activities in a patient who developed hyperthyroidism due to Graves’ disease following primary hypothyroidism. Clin Endocrinol (Oxf). 1986;25(5):519-25. 10. McDermott MT, Kidd GS, Dodson LE, Hofeldt FD. Hyperthyroidism following hypothyroidism. Am J Med Sci. 1986;291(3):194-8. 11. Starrenburg-Razenberg AJ, Castro Cabezas M, Gan IM, Njo TL, Rietveld AP, Elte JW. Four patients with hypothyroid Graves’ disease. Neth J Med. 2010; 68(4):178-80. 12. Leschik JJ, Diana T, Olivo PD, König J, Krahn U, Li Y, et al. Analytical performance and clinical utility of a bioassay for thyroid-stimulating immunoglobulins. Am J Clin Pathol. 2013;139(2):192-200. 13. McLachlan SM, Rapoport B. Thyrotropin-blocking autoantibodies and thyroid-stimulating autoantibodies: potential mechanisms involved in the pendulum swinging from hypothyroidism to hyperthyroidism or vice versa. Thyroid. 2013;23(1):14-24. 14. Elecsys TSH. Roche Diagnostics. Last updated March 2017. Available at: http://193.191.178.147/Mithras/Analyses.nsf/ecbed47964d bd5b9c1256cc6003eb951/410d3b08f3f0bffac1256c22002613bf/$FI LE/TSH-2-e8-201703.pdf. 15. Elecsys FT4II. Roche Diagnostics. Last uptdated March 2027. Available at: http://193.191.178.147/Mithras/Analyses.nsf/ecbed4 7964dbd5b9c1256cc6003eb951/9239e9f7863c35cec1256c220029 b5ed/$FILE/FT4II-2-e8-201702.pdf. 16. Elecsys FT3III. Roche Diagnostics. Last Updated March 2017. Available at: http://193.191.178.147/Mithras/Analyses.nsf/ecbed4 7964dbd5b9c1256cc6003eb951/8d82289784a0d256c1256c22002e 91a6/$FILE/FT3-2-e8-201706.pdf. 17. Anti TPO. Roche Diagnostics. Last Updated July 2017. Available at: http://labogids.sintmaria.be/sites/default/files/files/antitpo_2017-07_v5.pdf.

20. Leschik JJ, Diana T, Olivo PD, König J, Krahn U, Li Y, et al. Analytical performance and clinical utility of a bioassay for thyroid-stimulating immunoglobulins. Am J Clin Pathol. 2013;139(2):192-200. 21. Takasu N, Matsushita M. Changes of TSH-stimulation blocking antibody (TSBAb) and thyroid stimulating antibody (TSAb) over 10 years in 34 TSBAb-positive patients with hypothyroidism and in 98 TSAb-positive Graves’ patients with hyperthyroidism: reevaluation of TSBAb and TSAb in TSH-receptor-antibody (TRAb)positive patients. J Thyroid Res. 2012:182176. 22. Trifanescu R, Poiana C, Hortopan D. Autoinmune thyroid diseasea continuous spectrum. Rom J Intern Med. 2008; 46(4):361-5. 23. Takeda K, Takamatsu J, Kasagi K, Sakane S, Ikegami Y, Isotani H, et al. Development of hyperthyroidism following primary hypothyroidism: a case report with changes in thyroid-related antibodies. Clin Endocrinol. 1988;28(4):341-4. 24. Cho BY, Shong YK, Lee HK, Koh C-S, Min HK. Graves’ hyperthyroidism following primary hypothyroidism: sequential changes in various activities of thyrotropin receptor antibodies. Acta Endocrinol (Copenh). 1989;120:447-50. 25. Takasu N, Yamada T, Sato A, Nakagawa M, Komiya I, Nagasawa Y, et al. Graves’ disease following hypothyroidism due to Hashimoto’s disease: studies of eight cases. Clin Endocrinol (Oxf). 1990;33(6):687-98. 26. Kraiem Z, Baron E, Kahana L, Sadeh O, Sheinfeld M. Changes in stimulating and blocking TSH receptor antibodies in a patient undergoing three cycles of transition from hypo to hyperthyroidism and back to hypothyroidism. Clin Endocrinol (Oxf). 1992;36(2):211-4. 27. Dedecjus M, Stasiolek M, Brzezinski J, Selmaj K, Lewinski A. Thyroid hormones influence human dendritic cells’ phenotype, function, and subsets distribution. Thyroid. 2011;21(5):533-40. 28. Furqan S, Haque N, Islam N. Conversion of autoimmune hypothyroidism to hyperthyroidism. BMC Research Notes. 2014;7:489. 29. Schmidt M, Voell M, Rahlff I, Dietlein M, Kobe C, Faust M, et al. Long-term follow-up of antithyroid peroxidase antibodies in patients with chronic autoinmune thyroiditis (Hashimoto’s thyroiditis) treated with levothyroxine. Thyroid. 2008;18(7):755-60. 30. Dai YD, Carayanniotis G, Sercarz E. Antigen processing by autoreactive B cells promotes determinant spreading. Cell Mol Immunol. 2005;2(3):169-75. 31. Valdes-Socin H, Lutteri L, Cavallier E, Polus M, Geenen V, Louis E, et al. The thyrogastric autoimmune syndrome: its effects on micronutriments and gastric tumorigenesis. Rev Med Liege. 2013;68(11):579-84. 32. Valdes-Socin H, Sid S, Lutteri L, Polus M , Louis E , Beguin Y, et al. Le Syndrome Thyro-gastrique auto-immun: actualités cliniques et thérapeutiques. Vaisseaux, Cœur, Poumons. 2016;21(4) :40-5. 33. Aversa T, Lombardo F, Corrias A, Salerno M, De Luca F, Wasniewska M. In young patients with Turner or Down syndrome, Graves’ disease presentation is often preceded by Hashimoto’s thyroiditis. Thyroid. 2014;24(4):744-7.

Copyright© AE&M all rights reserved.

18. Elecsys AntiTSHR. Roche Diagnostics. Last Updated April 2017. Available at: http://www.cobas.com/home/product/clinical-andimmunochemistry-testing/elecsys-anti-tshr.html.

19. Osorio-Salazar C, Lecomte P, Madec AM, Baulieu JL. Basedow disease following autoinmune primary hypothyroidism. A propos of 7 cases. Ann Endocrinol. 1994;55(5):185-9.

614

Arch Endocrinol Metab. 2018;62/6


original article

Sex effects on the association between sarcopenia EWGSOP and osteoporosis in outpatient older adults: data from the SARCOS study Alberto Frisoli Jr.1,2, Fabíola Giannattasio Martin1,2, Antonio Carlos de Camargo Carvalho2, Jairo Borges1,2, Angela T. Paes3, Sheila Jean McNeill Ingham2,4

ABSTRACT

INTRODUCTION

S

arcopenia and osteoporosis are musculoskeletal clinical syndromes related to ageing, and are major public health concerns due to their likely bad outcomes. While the loss of bone mass increases the risk of fractures, loss of muscle mass and strength are strongly associated with a higher chance of falls (1,2). In turn, falls and fractures may lead to immobilization and a decrease in physical activity and these raise the odds of disability, hospitalization and the need for inpatient rehabilitation (3,4). Studies have demonstrated the correlation between low muscle mass and muscle strength and low bone mineral density in postmenopausal women (5) but not in men (6). However, the effect of bone on muscle mass and muscle strength has also been documented. Arch Endocrinol Metab. 2018;62/6

Correspondence to: Alberto Frisoli Jr. Av. das Nações Unidas, 14401 Torre Tarumã, cj. 2301 04794-000 – São Paulo, SP, Brasil frisoli@uol.com.br Received on May/13/2018 Accepted on Oct/3/2018 DOI: 10.20945/2359-3997000000087:

Juffer and cols. (7) has shown that osteocytes stimulated by mechanostatic action produce a number of factors, such as IGF-I, MGF, VEGF, and HGF which stimulate muscular function and formation. More recently, Yoshimura and cols. (8) showed that osteoporosis predicts incidental sarcopenia by the Asian Work Group for Sarcopenia (AWGS) in 4 years, but contrary to expectations, the opposite relationship was not significant. These results presented great variation due to the diverse concepts of sarcopenia, as well as due to uncontrolled variables that do not permit the establishment of a clear association between sarcopenia and osteoporosis with old age and gender. Based on these findings, we hypothesized that sarcopenia, by EWGSOP (European Working Group on Sarcopenia in Older People), has a strong association with osteoporosis 615

Copyright© AE&M all rights reserved.

Keywords Bone aging; muscle; body composition

Seção de Cardiogeriatria, Disciplina de Cardiologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil 2 Disciplina de Cardiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPMUnifesp), São Paulo, SP, Brasil 3 Departamento de Estatística, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil 4 Medicina Física e Reabilitação, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, SP, Brasil 1

Objective: The objective was to evaluate the association between sarcopenia (EWGSOP) and osteoporosis in older adults. Subjects and methods: This is a cross sectional analysis of a baseline evaluation of the SARCopenia and OSteoporosis in Older Adults with Cardiovascular Diseases Study (SARCOS). Three hundred and thirty-two subjects over 65 years of age were evaluated. Sarcopenia was determined by EWGSOP flowchart and Osteoporosis was established by WHO’s criteria. Physical function, comorbidities and medications were evaluated. Results: Women were older (79.8 ± 7.2 years) than men (78.21 ± 6.7 years) (p = 0.042). Osteoporosis occurred in 24.8% of men, and in 42.7% of women (p < 0.001); sarcopenia occurred in 25.5% of men and in 17.7%, of women (p = 0.103). Osteoporosis was diagnosed in 68% of sarcopenic women, however only 20.7% (p = 0.009) of women with osteoporosis had sarcopenia; in older men, 44.7% of individuals with sarcopenia presented osteoporosis and 42.9% (p = 0.013) of men with osteoporosis showed sarcopenia. In an adjusted logistic regression analyses for sarcopenia, osteoporosis presented a statistically significant association with sarcopenia in men [OR: 2.930 (95% CI: 1.044-8.237; p = 0.041)] but not in women [OR: 2.081 (0.787-5.5; p = 0.142)]; in the adjusted logistic regression analyses for osteoporosis, a statistically significant association occurred in men [OR: 2.984 (95% CI: 1.144-7.809; p = 0.025)], but not in women [OR: 2.093 (0.962-3.714; p = 0.137)]. Conclusion: According to sex, there are significant differences in the association between sarcopenia EWGSOP and osteoporosis in outpatient older adults. It is strong and significant in males; in females, despite showing a positive trend, it was not statistically significant. Arch Endocrinol Metab. 2018;62(6):615-22


Osteoporosis and sarcopenia in elderly

in older females but not in older males; additionally, we believe that the inverse association, i.e. osteoporosis with sarcopenia by EWGSOP will show the same trend in both sexes. To test our hypothesis, we evaluated the association between osteoporosis according to the WHO’s definition (9) and sarcopenia according to the EWGSOP (10) in older men and women from the same population group, with established risk factors and significant variables that could interfere with bone mineral density, muscle mass and muscle strength.

Bone mineral density and osteoporosis

SUBJECTS AND METHODS

Bone mineral density (BMD – g/cm2) of the lumbar spine, femoral neck, total femur and appendicular muscle mass and total fat mass were assessed through a DXA analysis by dual-energy X-ray absorptiometry (GE Lunar; DPX-MD 73477, GE Medical system, Madison, WI). Osteoporosis was established by the WHO’s criteria (9), i.e., BMD T score ≤ -2.5 standard deviations (SD) at lumbar spine, femur neck, and total femur.

Subjects

Disability

This study is a cross sectional analysis of a baseline evaluation of the SARCopenia and OSteoporosis in Older Adults with Cardiovascular Diseases Study (SARCOS), a one-year prospective cohort study that investigated the association between cardiovascular diseases and changes in body composition, muscle strength and physical performance as a common pathway to disability. We interviewed 383 older outpatient adults from an outpatient cardio-geriatric clinic and 332 were included in this study and underwent DXA analyzes. Our population was composed by older adults, over 65 years of age, both sexes and all ethnic groups. Exclusion criteria were: unstable medical conditions, any form of cancer in the last five years, chronic renal failure in dialysis, Parkinson’s disease, severe infectious disease requiring hospitalization in the previous month, moderate or severe dementia classified by the MMSE (mini-mental state examination) (11,12) and use of gait assistant devices. This study was approved by the Ethical Review Board at our Institution and written informed consent was obtained from all participants.

Disability was assessed by the number of tasks performed in activities of daily living (ADL) and instrumental activities of daily living (IADL); the cut point for disability was 5 for ADL or 25 for IADL (13-15).

Diagnosis of sarcopenia

Copyright© AE&M all rights reserved.

a handheld dynamometer (Jamar; TEC, Clifton, NJ, USA); maximum values are reported.

Sarcopenia, as determined by EWGSOP’s flowchart (10), is defined by the presence of weakness represented by grip strength of the dominant hand lower than 20 kgf for women and 30 kgf for men and/or a gait speed lower than 0.8 m/s, plus low appendicular muscle mass by height2 lower than 5.45 kg/m2 for women and 7.26 kg/m2 for men.

Handgrip strength Isometric grip strength of the dominant upper extremity was determined by three measurements with 616

Other measurements Demographic data, weight, height, cardiovascular disease (arterial hypertension, atrial fibrillation, previous myocardial infarction, chronic atherosclerosis coronary, heart failure, peripheral arterial obstruction) and other chronic diseases: diabetes mellitus osteoarthritis, nondialysis dependent chronic kidney disease (CKD), chronic obstructive pulmonary disease (COPD), previous diagnosis of cancer, current or previous consumption of alcohol (at least one year without drinking alcohol), and current or previous smoking history (at least five years without smoking). We also considered falls (at least one fall in last 6 months) and history of fractures (a clinical fracture or diagnosed by radiograph assessment in a Health Care service). Finally, we evaluated all medications that could interfere in bone and muscle metabolism, such as bisphosphonates, tereparatide, strontium ranelate, corticosteroids in high doses (≥ 7.5 mg predinisone/day or equivalent for more than 3 months), vitamin D over 800 IU/ day, estrogen and progesterone replacements, ACE (angiotensin-converting-enzyme) inhibitors, ARB (angiotensin II receptor blocker) I and II inhibitors.

Statistical analysis Qualitative variables are expressed as absolute and relative frequencies. Quantitative data are summarized as means, medians, standard deviations, minimum and maximum values. To compare the groups (sarcopenia Arch Endocrinol Metab. 2018;62/6


Osteoporosis and sarcopenia in elderly

and osteoporosis) the chi-square test for qualitative variables and ANOVA for quantitative variables were used to compare differences in baseline characteristics by gender. Binomial logistic regression analyses were performed to evaluate the association between osteoporosis (OP) with sarcopenia EWGSOP and vice versa. Adjusted regression analyses were performed with significant variables for sarcopenia (current use of ACE inhibitor/ ARB I and II inhibitors, falls, previous fracture (only for women), diabetes mellitus and disability), and for osteoporosis (falls, age, previous alcohol consumption, disability, smoking history, bisphosphonate use and

diabetes mellitus). In the case of two quantitative variables, scatter diagrams and correlation coefficients were used. SPSS version 22 (SPSS, Inc., Chicago, IL, USA) statistical software package was used for carrying out all the analyses. Statistical significance was set at 0.05.

RESULTS Demographic data, body composition parameters, muscle strength, prevalence of chronic and cardiovascular diseases, osteoporosis, and sarcopenia are described in Table 1.

Table 1. Demographic data, body composition parameters, muscle strength, prevalence of chronic and cardiovascular diseases, osteoporosis, and sarcopenia EWGSOP All (N = 332)

Men (N = 141)

Women (N = 191)

P value

Age (years) (average (SD))

78.44 (7.16)

78.21 (6.78)

79.81 (7.24)

0.042

Years of education (average (SD))

3.52 (3.03)

4.54 (3.51)

3.48 (2.94)

0.003

Personal income (average (SD))

1.63 (1.79)

2.13 (2.50)

1.24 (0.90)

< 0.001

Number of medications (average (SD))

6.48 (2.61)

6.20 (2.29)

6.65 (2.80)

0.124

Grip strength (kgf) (average (SD))

22.56 (7.86)

28.58 (6.49)

17.63 (4.78)

< 0.001

Lumbar spine BMD (g/cm2) (average (SD))

1.075 (0.223)

1.182 (0.22)

0.993 (0.20)

< 0.001

Femur neck BMD (g/cm ) (average (SD))

0.830 (0.154)

0.883 (0.16)

0.781 (0.13)

< 0.001

Total femur BMD (g/cm ) (average (SD))

0.871 (0.168)

0.931(0.16)

0.806 (0.14)

< 0.001

Total body fat (%) (average (SD))

39.62 (9.62)

33.88 (7.75)

44.16 (8.48)

< 0.001

IAMM (kg/m ) (average (SD))

6.74 (3.33)

7.21 (0.84)

6.00 (0.86)

< 0.001

Hypertension (%)

92.1

92.2

92.0

0.832

Diabetes mellitus (%)

39.8

39.5

42.2

0.713

Previous consumption of alcohol (%)

16.3

33.9

2.6

< 0.001

Myocardial infarction (%)

32.6

42.7

26.9

0.007

Heart failure (%)

28.9

28.2

30.3

0.791

Previous diagnosis of osteoporosis (%)

20.9

6.5

31.4

< 0.001

COPD (%)

8.4

10.7

6.7

0.194

Previous fracture (%)

31.4

33.1

30.1

0.602

CKD (%)

16.3

16.9

14.8

0.743

Osteoarthritis (%)

37.3

22.0

46.8

< 0.001

Smoking history (%)

49.7

69.4

32.1

< 0.001

Disability

44.8

57.1

42.9

0.548

Falls in last 6 months (%)

27.1

16.3

30.8

0.007

Stroke (%)

15.4

15.3

16.0

1.061

18.4

25.5 (36)

17.7 (28)

0.103

24.8 (35)

42.7 (82)

< 0.001

2

2

2

Present diagnosis of sarcopenia and osteoporosis Sarcopenia EWGSOP (%) Osteoporosis (OP) (%)

36

Personal income: US$ 312.50/month; IAMM: index of appendicular muscle mass (AMM/height ). Sarcopenia by EWGSOP (European Working Group on Sarcopenia in Older People); OP: osteoporosis at proximal femur and/or lumbar spine. COPD: chronic obstructive pulmonary disease; CKD: chronic kidney disease. P values refer to the difference between men and women. 2

Arch Endocrinol Metab. 2018;62/6

617

Copyright© AE&M all rights reserved.

Prior diagnosis of chronic diseases


Osteoporosis and sarcopenia in elderly

Prevalence of osteoporosis and sarcopenia The characteristics of the older adults with and without osteoporosis and sarcopenia are shown in Tables 2 and 3.

The association of osteoporosis and sarcopenia EWGSOP in older men and women Osteoporosis was diagnosed in 52.5% (n = 32; p = 0.002) of subjects with sarcopenia, but sarcopenia was only diagnosed in 27.4% of subjects with osteoporosis. This trend was observed in women, where 68% of sarcopenic patients showed osteoporosis, and only 20.7% (n = 17; p = 0.009) of osteoporotic patients showed sarcopenia; contrary to this, 44.7% of

men with sarcopenia presented with osteoporosis and 42.9%; (n = 15; p = 0.013) of men with osteoporosis presented with sarcopenia. Both disorders occurred in 10.6% of men and 8.9% of women. In the logistic regression analyses for sarcopenia, osteoporosis presented a similar value of the association for men OR=3.03 (95% CI: 1.334-6.909; p = 0.008) and women OR = 3.30 (95%CI: 1.347-8.091; p = 0.009), and vice versa. In the adjusted logistic regression analyses, for sarcopenia and osteoporosis, we used different variables for, for men and women, according to statistical significance that they had presented previously (Tables 2 and 3). Variables used in

Table 2. Demographic data and prevalence of chronic and cardiovascular diseases, in men and women with and without osteoporosis Men

Women

Non-Osteoporosis

Osteoporosis

p

Non-Osteoporosis

Osteoporosis

p

Age (years) (average (SD))

77.3 (6.4)

81.8 (7.3)

< 0.001

78.7 (6.6)

80.9 (7.8)

0.049

Caucasians (%)

60.4

71.9

0.405

72.6

71.2

0.377

Smoking history (%)

67.2

65.4

1.054

23.4

33.9

0.282

Previous consumption of alcohol (%)

31.1

42.3

0.331

0

5.4

0.241

Falls in last 6 months (%)

Subject characteristics

16.4

19.2

0.764

27.7

26.8

1.003

Lumbar spine BMD (g/cm2) (average (SD))

1.230 (0.20)

0.992 (0.15)

< 0.001

1.119 (0.15)

0.844 (0.12)

< 0.001

Femur neck BMD (g/cm2) (average (SD))

0.935 (0.14)

0.735 (0.10)

< 0.001

0.870 (0.10)

0.683 (0.10)

< 0.001

Total femur BMD (g/cm2) (average (SD))

0.990 (0.14)

0.781 (0.12)

< 0.001

0.906 (0.11)

0.705 (0.11)

< 0.001

IAMM (kg/m2) (average (SD))

7.376 (0.88)

6.704 (0.93)

< 0.001

6.263 (0.94)

6.571 (6.52)

0.627

Total body fat (%) (average (SD))

34.78 (7.22)

31.00 (8.95)

0.009

45.36 (7.61)

42.14 (9.18)

0.008

29.4 (6.5)

26.2 (6.6)

0.022

18.5 (4.7)

16.7 (5.0)

0.027

Hypertension (%)

92.3

84.4

0.296

91.6

90.4

1.000

Diabetes mellitus (%)

47.3

18.8

0.006

48.2

34.7

0.104

Heart failure (%)

25.3

37.5

0.254

66.3

26.0

0.382

Osteoarthritis (%)

23.3

18.8

0.632

47.6

45.2

0.873

Previous stroke (%)

13.2

18.8

0.561

14.3

17.8

0.663

Chronic kidney disease (%)

16.5

18.8

0.788

15.7

13.7

0.823

Previous cancer (%)

14.3

28.1

0.107

14.3

6.80

0.198

Disability (%)

37.2

48.7

0.256

38.4

60.5

0.003

History of osteoporosis (%)

3.3

15.6

0.028

27.4

35.6

0.302

Previous fractures (%)

27.5

50.0

0.029

27.4

32.9

0.488

Dominant grip strength (kgf) (average (SD))

CopyrightŠ AE&M all rights reserved.

Comorbidities

Note: P values refer to the difference between men and women with and without osteoporosis. IAMM: index of appendicular muscle mass (AMM/height2).

618

Arch Endocrinol Metab. 2018;62/6


Osteoporosis and sarcopenia in elderly

the logistic regression analyses for sarcopenia in the female group were: age, previous clinical fractures, diabetes mellitus, falls in the last 6 months, cancer history, ACE/ ARB I and II use and disability; for the male group, the same variables were used, with the exception of previous clinical fracture and cancer history. In the osteoporosis analyzes, the variables used in the female group were: age, smoking history, diabetes mellitus, falls in the last 6 months, previous consumption of alcohol, current use of bisphosphonates and disability; for the male group the same variables were used, with the exception of disability. After the adjustment, osteoporosis presented a significant association with sarcopenia only in men with an OR: 2.930, (95% CI: 1.04-8.23; p = 0.041) and this trend remained in the analyses for osteoporosis, where

sarcopenia presented an OR: 2.984 (1.144-7.809; p = 0.025). While in women, despite the analysis showing a positive trend in the association between osteoporosis and sarcopenia (OR: 2.081 (0.787-5.5; p = 0.142)), and vice versa (OR: 2.093 (0.962-3.714; p = 0.137)) they did not reach statistical significance (Table 4). Interestingly, contrary to the previous literature (10,16,17), age was not an independent predictor of osteoporosis and sarcopenia, in both sexes. Diabetes mellitus was negatively associated with osteoporosis in men, but in women this association was found to be inverse and also significant (Table 4). Disability showed the highest association with sarcopenia in both genders in comparison with the other variables. Finally, in women, previous fractures were also associated with sarcopenia (Table 4).

Table 3. Demographic data, body composition parameters, muscle strength, prevalence of chronic and cardiovascular diseases, of men and women with and without sarcopenia EWGSOP Men NonSarcopenia

Sarcopenia

Age (years old) (average (SD))

76.90 (6.20)

Caucasians (%) Smoking history (%) Previous consumption of alcohol (%)

Women p

NonSarcopenia

Sarcopenia

p

82.91 (7.00)

< 0.001

79.02 (6.95)

82.64 (8.06)

0.016

60.4

71.9

0.059

70.8

78.6

0.331

70.3

69.4

1.002

33.5

36.0

0.822

31.4

33.3

0.865

3.4

4.0

1.009

Subject characteristics

Falls in last 6 months (%)

17.8

34.3

0.050

27.9

48.0

0.062

Lumbar spine BMD (g/cm2) (average (SD))

1.185 (0.22)

1.139 (0.21)

0.238

1.014 (0.19)

0.926 (0.23)

0.043

Femur neck BMD (g/cm ) (average (SD))

0.915 (0.15)

0.822 (0.16)

0.003

0.804 (0.13)

0.686 (0.10)

< 0.001

Total Femur BMD (g/cm2) (average (SD))

0.971 (0.16)

0.873 (0.17)

0.002

0.839 (0.14)

0.709 (0.12)

< 0.001

2

IAMM (kg/m ) (average (SD))

7.57 (0.80)

6.24 (0.64)

< 0.001

6.62 (4.63)

4.97 (0.36)

0.076

Total body fat (%) (average (SD))

33.82 (7.46)

33.48 (8.67)

0.825

44.37 (8.34)

40.97 (9.36)

0.063

Dominant grip strength (kgf) (average (SD))

31.08 (5.27)

21.41 (4.76)

< 0.001

18.67 (4.58)

12.92 (3.28)

< 0.001

89

93.8

0.512

91.1

86.1

0.512

Diabetes mellitus (%)

45.1

25.0

0.431

45.6

22.2

0.022

Heart Failure (%)

29.7

25.0

0.654

27.8

33.3

0.660

Osteoarthritis (%)

22.2

21.9

1.010

23.1

16.7

0.473

Previous stroke (%)

13.2

18.8

0.562

12.7

22.2

0.267

Chronic kidney disease (%)

15.4

21.9

0.422

15.2

16.7

1.000

Previous cancer (%)

14.3

28.1

0.101

13.9

30.6

0.043

Disability (%)

31.4

77.8

< 0.001

40.8

84.0

< 0.001

History of osteoporosis (%)

5.5

9.4

0.687

31.5

28.6

0.821

Previous fractures (%)

30.8

40.6

0.383

25.3

47.2

0.031

2

Hypertension (%)

Note: P values refer to the difference between men and women with and without osteoporosis. IAMM: index of appendicular muscle mass (AMM/height2). Arch Endocrinol Metab. 2018;62/6

619

CopyrightŠ AE&M all rights reserved.

Comorbidities


Osteoporosis and sarcopenia in elderly

Table 4. Adjusted logistic regression analyses for sarcopenia EWGSOP in older women and men Sarcopenia EWGSOP in Men OR (95% CI; p)

Sarcopenia EWGSOP in Women OR (95% CI; p)

Osteoporosis

2.930 (1.044-8.237; p = 0.041)

2.081 (0.787-5.5; p = 0.142)

Age

1.053 (0.98-1.132; p = 0.161)

1.051 (0.982-1.125; p = 0.151)

Previous fractures

0.894 (0.328-2.441; p = 0.828)

Diabetes mellitus

2.462 (0.926-6.549; p = 0.071)

0.971 (0.36-2.621; p = 0.954)

Falls in last 6 months

1.635 (0.585-4.567; p = 0.349)

2.164 (0.85-5.511; p = 0.105)

Cancer history

0.467 (0.054-4; p = 0.487)

ACE inhibitor/ARB I and II inhibitors current use

1.333 (0.516-3.442; p = 0.553)

0.597 (0.211-1.684; p = 0.329)

Disability

6.546 (2.476-17.273; p < 0.001)

4.904 (1.487-16.172; p = 0.009)

Note: Adjusted logistic regression – variables used in the female group were: age, previous clinical fractures, diabetes mellitus, falls in the last 6 months, cancer history, ACE/ARB I and II use and disability; for the male group, the same variables were used, with the exception of previous clinical fracture and cancer history.

Copyright© AE&M all rights reserved.

DISCUSSION To the best of our knowledge, this is the first study to demonstrate that there are important differences in the association between sarcopenia EWGSOP and osteoporosis in older adults determined by sex. Contrary to our hypothesis, females did have an association between osteoporosis and sarcopenia EWGSOP; however, this association did not remain relevant after adjustments for confounder clinical variables were performed. Previous studies (18,19) on the relationship between loss of muscle mass and strength and osteoporosis have shown great variation according to sex, age and health, diagnostic criteria for sarcopenia and cutoff points used for bone loss; these factors may cause a significant variation in the association values. One of the few studies that used the EWGSOP’s criteria analyzed 409 independent women, aged 70-80 years from the community, and did not find a significant association between low BMD and sarcopenia (20) although the prevalence of sarcopenia by EWGSOP was very low (0.9%) and only 36% of those women presented with osteopenia. Our findings in women differ significantly from others, but the population, the definition of sarcopenia, and confounder variables evaluated were different. In the Osteoporosis Risk Factor and Prevention (OSTPRE) Study, women with sarcopenia by EWGSOP had 12.9 times (3.1–53.5; p < 0.001) higher odds of having osteoporosis when compared to women without sarcopenia; but in the OSTPRE study the sample was composed by younger (68.7 ± 1.8 yo) postmenopausal women from the dwelling community. Besides, in our outpatient population, 620

sarcopenia diagnosis was made with a higher cut off for lean mass (cut-off of 6.3 kg/m2) (21). Another key point of our data was the diversity in the correlation between diabetes and osteoporosis among the sexes. In older women, the presence of diabetes has shown a higher risk of osteoporosis, whereas in men it appears to have a protective effect. However, these results should be evaluated with caution, since the study was not designed for this purpose. Higher levels of BMD in men with diabetes compared to non–diabetes subjects were also described in the Rotterdam, EVOS and The Health ABC studies; they have demonstrated 3-5% higher bone site BMD in men with diabetes vs. nondiabetes (22-24). The greater tendency of osteoporosis in women, evidenced in our series and also present in other studies, may be justified by the earlier estrogenic deprivation caused by menopause, by the other hormone deficiencies, more comorbidities (25,26), and by the process of inflammaging (27). In men, osteoporosis usually begins during the seventh decade (28) justifying the difference in prevalence between sexes due to the decrease of testosterone. Estrogen deprivation, also affects the incidence of sarcopenia, but mainly by the loss of muscle strength (29). This theory is endorsed by studies noting that muscle strength is preserved in women who opt for hormone replacement therapy at the onset of menopause, as compared with those who do not (30). We believe that screening for osteoporosis in older adult outpatients should be recommended not only to evaluate the risk of fractures through the analysis of BMD but additionally, to evaluate the risk of sarcopenia, an important risk factor for falls and Arch Endocrinol Metab. 2018;62/6


Osteoporosis and sarcopenia in elderly

2. Gonzalez-Montalvo JI, Alarcon T, Gotor P, Queipo R, Velasco R, Hoyos R, et al. Prevalence of sarcopenia in acute hip fracture patients and its influence on short-term clinical outcome. Geriatr Gerontol Int. 2016;16(9):1021-7. 3. Craig J, Murray A, Mitchell S, Clark S, Saunders L, Burleigh L. The high cost to health and social care of managing falls in older adults living in the community in Scotland. Scott Med J. 2013;58(4):198-203. 4. Rapp K, Rothenbacher D, Magaziner J, Becker C, Benzinger P, Konig HH, et al. Risk of Nursing Home Admission After Femoral Fracture Compared With Stroke, Myocardial Infarction, and Pneumonia. J Am Med Dir Assoc. 2015;16(8):715.e7-715.e12. 5. Clynes MA, Edwards MH, Buehring B, Dennison EM, Binkley N, Cooper C. Definitions of Sarcopenia: Associations with Previous Falls and Fracture in a Population Sample. Calcif Tissue Int. 2015;97(5):445-52. 6. Ribom E, Ljunggren O, Piehl-Aulin K, Ljunghall S, Bratteby LE, Samuelson G, et al. Muscle strength correlates with total body bone mineral density in young women but not in men. Scand J Med Sci Sports. 2004;14(1):24-9. 7.

Juffer P, Jaspers RT, Lips P, Bakker AD, Klein-Nulend J. Expression of muscle anabolic and metabolic factors in mechanically loaded MLO-Y4 osteocytes. Am J Physiol Endocrinol Metab. 2012;302(4):E389-95.

8. Yoshimura N, Muraki S, Oka H, Iidaka T, Kodama R, Kawaguchi H, et al. Is osteoporosis a predictor for future sarcopenia or vice versa? Four-year observations between the second and third ROAD study surveys. Osteoporos Int. 2017;28(1):189-99. 9. World Health Organization. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Geneve: WHO technical report series; 1994. 10. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412-23. 11. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-98. 12. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56(3):M146-56. 13. Katz S, Downs TD, Cash HR, Grotz RC. Progress in development of the index of ADL. Gerontologist. 1970;10(1):20-30. 14. Katz S, Ford AB, Moskowitz RW, Jackson BA, Jaffe MW. Studies of Illness in the Aged. The Index of Adl: A Standardized Measure of Biological and Psychosocial Function. JAMA. 1963;185:914-9.

Study was performed at: School of Medicine, Federal University of São Paulo, São Paulo, Brazil.

15. Lawton MP, Brody EM. Assessment of older people: self-maintaining and instrumental activities of daily living. Gerontologist. 1969;9(3):179-86.

Funding: this research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

16. Baumgartner RN, Koehler KM, Gallagher D, Romero L, Heymsfield SB, Ross RR, et al. Epidemiology of sarcopenia among the elderly in New Mexico. Am J Epidemiol. 1998 Apr 15;147(8): 755-63.

Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1.

Hida T, Ishiguro N, Shimokata H, Sakai Y, Matsui Y, Takemura M, et al. High prevalence of sarcopenia and reduced leg muscle mass in Japanese patients immediately after a hip fracture. Geriatr Gerontol Int. 2013;13(2):413-20.

Arch Endocrinol Metab. 2018;62/6

17. Studenski SA, Peters KW, Alley DE, Cawthon PM, McLean RR, Harris TB, et al. The FNIH sarcopenia project: rationale, study description, conference recommendations, and final estimates. J Gerontol A Biol Sci Med Sci. 2014;69(5):547-58. 18. Verschueren S, Gielen E, O’Neill TW, Pye SR, Adams JE, Ward KA, et al. Sarcopenia and its relationship with bone mineral density in middle-aged and elderly European men. Osteoporos Int. 2013;24(1):87-98. 19. Rikkonen T, Sirola J, Salovaara K, Tuppurainen M, Jurvelin JS, Honkanen R, et al. Muscle strength and body composition are clinical indicators of osteoporosis. Calcif Tissue Int. 2012;91(2):131-8.

621

Copyright© AE&M all rights reserved.

fractures. This strong association between osteoporosis and sarcopenia, and vice versa, especially in older men, must be considered when deciding upon therapeutic strategies for the prevention of fractures, since it is of utmost importance that both conditions are treated. This study has limitations. First, our sample size is small, although it is considerable if we concede that it is a very old population (average age 80 yo). Another limitation of our study was the non-radiological confirmation of bone fractures of the majority of patients who reported a history of fracture, which may cause a bias on the analyses of the osteoporotic sample, since previous fractures should be considered as having osteoporosis, independent of DXA. Also, as the average age is high, we cannot extrapolate our findings to a younger population. This study is a cross sectional analyses and, as such, does not allow us to establish a cause and effect relationship between the loss of BMD and the loss of appendicular muscle mass and/or muscle strength. We did not quantify myokines that could help shed some light on the interaction between loss of bone and muscle mass/strength. In summary, older adults from an outpatient clinic setting presented with a high prevalence of osteoporosis and/or sarcopenia EWGSOP. In regards to sex, sarcopenia EWGSOP was more prevalent in males while osteoporosis was more prevalent in females. The interaction between muscle mass and muscle function with bone metabolism seems to be more intense in older men than in older women. In conclusion, according to sex, there are significant differences in the association between sarcopenia EWGSOP and osteoporosis in outpatient older adults. It is strong and significant in males; in females, despite showing a positive trend, it was not statistically significant.


Osteoporosis and sarcopenia in elderly

20. Patil R, Uusi-Rasi K, Pasanen M, Kannus P, Karinkanta S, Sievanen H. Sarcopenia and osteopenia among 70-80-year-old homedwelling Finnish women: prevalence and association with functional performance. Osteoporos Int. 2013;24(3):787-96.

25. Reginster JY, Burlet N. Osteoporosis: a still increasing prevalence. Bone. 2006;38(2 Suppl 1):S4-9.

21. Sjoblom S, Suuronen J, Rikkonen T, Honkanen R, Kroger H, Sirola J. Relationship between postmenopausal osteoporosis and the components of clinical sarcopenia. Maturitas. 2013;75(2):175-80.

27. De Martinis M, Di Benedetto MC, Mengoli LP, Ginaldi L. Senile osteoporosis: is it an immune-mediated disease? Inflamm Res. 2006;55(10):399-404.

22. van Daele PL, Stolk RP, Burger H, Algra D, Grobbee DE, Hofman A, et al. Bone density in non-insulin-dependent diabetes mellitus. The Rotterdam Study. Ann Intern Med. 1995;122(6):409-14.

28. Puth MT, Klaschik M, Schmid M, Weckbecker K, Munster E. Prevalence and comorbidity of osteoporosis- a cross-sectional analysis on 10,660 adults aged 50 years and older in Germany. BMC Musculoskelet Disord. 2018;19(1):144.

23. Lunt M, Masaryk P, Scheidt-Nave C, Nijs J, Poor G, Pols H, et al. The effects of lifestyle, dietary dairy intake and diabetes on bone density and vertebral deformity prevalence: the EVOS study. Osteoporos Int. 2001;12(8):688-98.

29. Greeves JP, Cable NT, Reilly T, Kingsland C. Changes in muscle strength in women following the menopause: a longitudinal assessment of the efficacy of hormone replacement therapy. Clin Sci (Lond). 1999;97(1):79-84. 30. Phillips SK, Rook KM, Siddle NC, Bruce SA, Woledge RC. Muscle weakness in women occurs at an earlier age than in men, but strength is preserved by hormone replacement therapy. Clin Sci (Lond). 1993;84(1):95-8.

CopyrightŠ AE&M all rights reserved.

24. Strotmeyer ES, Cauley JA, Schwartz AV, Nevitt MC, Resnick HE, Zmuda JM, et al. Diabetes is associated independently of body composition with BMD and bone volume in older white and black men and women: The Health, Aging, and Body Composition Study. J Bone Miner Res. 2004;19(7):1084-91.

26. Kanis JA, McCloskey EV. Risk factors in osteoporosis. Maturitas. 1998;30(3):229-33.

622

Arch Endocrinol Metab. 2018;62/6


original article

A pioneering RET genetic screening study in the State of Ceará, Brazil, evaluating patients with medullary thyroid cancer and at-risk relatives: experience with 247 individuals Maria Cecília Martins-Costa1,2,3, Susan C. Lindsey1, Lucas L. Cunha1, Fernando Porto Carreiro-Filho4, André P. Cortez5,6, Marcelo E. Holanda5, J. Wilson M. de Farias4,5, Sérgio B. Lima5, Luís A. Albano Ferreira7,8, Pedro Collares Maia Filho9, Cléber P. Camacho1, Gilberto K. Furuzawa1, Ilda S. Kunii1, Magnus R. Dias-da-Silva1, João R. M. Martins1,10, Rui M. B. Maciel1,11

ABSTRACT Objective: Initial diagnosis of medullary thyroid carcinoma (MTC) is frequently associated with advanced stages and a poor prognosis. Thus, the need for earlier diagnoses and detection in relatives at risk for the disease has led to increased use of RET genetic screening. Subjects and methods: We performed RET screening in 247 subjects who were referred to the Brazilian Research Consortium for Multiple Endocrine Neoplasia (BRASMEN) Center in the State of Ceará. Direct genetic sequencing was used to analyze exons 8, 10, 11, and 13-16 in MTC index cases and specific exons in at risk relatives. Afterward, clinical follow-up was offered to all the patients with MTC and their affected relatives. Results: RET screening was performed in 60 MTC index patients and 187 at-risk family members. At the initial clinical assessment of the index patients, 54 (90%) were diagnosed with apparently sporadic disease and 6 (10%) diagnosed with hereditary disease. After RET screening, we found that 31 (52%) index patients had sporadic disease, and 29 (48%) had hereditary disease. Regarding at-risk relatives, 73/187 were mutation carriers. Mutations in RET codon 804 and the rare p.M918V mutation were the most prevalent. Conclusions: Performing RET screening in Ceará allowed us to identify a different mutation profile in this region compared with other areas. RET screening also enabled the diagnosis of a significant number of hereditary MTC patients who were initially classified as sporadic disease patients and benefited their relatives, who were unaware of the risks and the consequences of bearing a RET mutation. Arch Endocrinol Metab. 2018;62(6):623-35

INTRODUCTION

M

edullary thyroid carcinoma (MTC) is an uncommon malignant tumor arising from the calcitonin-producing parafollicular cells (C-cells) of the thyroid and accounts for 1-2% of all thyroid cancers in the US (1). Regarding the Brazilian population, the latest estimates indicate that 5,870 Arch Endocrinol Metab. 2018;62/6

Correspondence to: Rui M. B. Maciel / João R. M. Martins Laboratório de Endocrinologia Molecular e Translacional, Escola Paulista de Medicina, Universidade Federal de São Paulo Rua Pedro de Toledo, 669, 11º andar 04039-32 – São Paulo, SP, Brasil rui.maciel@unifesp.br j.martins@unifesp.br Received on June/29/2018 Accepted on Sept/25/2018 DOI: 10.20945/2359-3997000000088:

and 1,090 new cases of all types of thyroid cancer have been diagnosed among women and men, respectively, in 2016 (2). Regarding Ceará, a State of the Northeastern region of Brazil with an estimated population of 9,107,101 inhabitants, the latest estimates indicate that 460 and 100 new cases of all types of thyroid cancer have been diagnosed among women and men, respectively (2). 623

Copyright© AE&M all rights reserved.

Keywords Medullary thyroid carcinoma; RET mutation; genetic screening

Centro de Doenças da Tiroide e Laboratório de Endocrinologia Molecular e Translacional, Divisão de Endocrinologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, SP, Brasil 2 Centro de Endocrinologia e Metabologia, Hospital Geral de Fortaleza (HGF), Fortaleza, CE, Brasil 3 Departamento de Medicina, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brasil 4 Departamento de Cirurgia de Cabeça e Pescoço, Hospital Geral de Fortaleza, Fortaleza, CE, Brasil 5 Santa Casa de Misericórdia de Fortaleza, Fortaleza, CE, Brasil 6 Hospital Geral Dr. César Cals, Fortaleza, CE, Brasil 7 Hospital Universitário Walter Cantídio, Universidade Federal do Ceará (UFC), Fortaleza, CE, Brasil 8 Hospital Infantil Albert Sabin, Fortaleza, CE, Brasil 9 Centro Universitário Christus (Unichristus), Fortaleza, CE, Brasil 10 Divisão de Biologia Molecular, Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, SP, Brasil 11 Fleury Medicina e Saúde, São Paulo, SP, Brasil 1


RET screening in the Northeast of Brazil

In a Brazilian study about thyroid cancer incidence patterns in the State of São Paulo from 1997-2008, Veiga and cols. noted incidences of MTC of 0.56 per 100,000 persons in women and 0.15 per 100,000 persons in men (3). Assuming that the incidence of this neoplasm in Ceará is similar to that reported in the State of São Paulo, approximately 26 and 7 new cases of MTC would be diagnosed in women and men, respectively, per year (3). The sporadic form of MTC is the most common presentation of MTC (approximately 75% of cases) (4), but in 25% of cases, MTC may also occur as part of an inherited disorder referred to as multiple endocrine neoplasia type 2 (MEN 2). MEN 2 is caused by germline mutations in the REarranged during Transfection (RET) gene and is transmitted as an autosomal dominant trait with variable degrees of expressivity and age-related penetrance (5). The clinical diagnosis of MTC by palpation of a thyroid nodule or mass on physical examination is usually associated with advanced TNM stages and a poor prognosis. In this context, RET molecular analysis assumes vital importance, as this analysis allows earlier diagnosis of MTC, thereby increasing the chance of curative treatment (1). Additionally, RET molecular analysis enables the diagnosis of hereditary MTC in some apparently sporadic cases, which favors more careful management of other diseases that may accompany MEN 2 syndrome. This study aimed to report the clinical pattern of MTC in the State of Ceará and to describe the molecular profile of this neoplasm using RET gene analysis.

SUBJECTS AND METHODS

Copyright© AE&M all rights reserved.

Subjects We report the results of our experience with RET genetic screening of patients with a diagnosis of MTC, as well as their at-risk relatives, who were evaluated at a single center in Ceará, Brazil, from May 2009 to December 2015. This center is located in the city of Fortaleza, the capital of Ceará, and is part of a Brazilian Research Consortium for Multiple Endocrine Neoplasia (BRASMEN). All RET sequencings performed in this study were conducted in the Laboratory of Molecular and Translational Endocrinology, Division of Endocrinology, Department of Medicine, Escola 624

Paulista de Medicina, Universidade Federal de São Paulo (EPM/Unifesp). All subjects investigated in this study signed informed consent. The study protocol was approved by the local internal review board (CAAE: 16441414.9.1001.5505111). The patients included in this study had histopathologically confirmed diagnoses of MTC. Immunohistochemical studies for calcitonin, synaptophysin, and chromogranin were performed when histopathological exams were inconclusive, and no patients presented with elevated serum calcitonin (sCt) or CEA levels. All the patients were originally from Ceará. The exclusion criteria were: patients from other countries or other Brazilian States, relatives of deceased MTC index patients without available histopathologic information to confirm their relative’s MTC diagnosis and patients with uncertain histopathology.

Clinical evaluation before RET screening Patients initially underwent a clinical assessment and genetic counseling before undergoing peripheral blood collection for RET analysis. Patients were warned about the risks of having additional endocrine diseases related to MEN 2 if a RET mutation was identified, as well as the risks facing their relatives bearing the same mutation. Therefore, a portion of the patient population analyzed in this study comprised the at-risk relatives of patients diagnosed with hereditary MTC after RET sequencing. In addition, a questionnaire was designed to acquire the following general information about each patient: age upon diagnosis of MTC/CCH and upon RET screening; previous knowledge of MTC/CCH upon total thyroidectomy (TT), if previously performed; preoperative cervical ultrasound or thyroid nodule or lymph node (LN) fine-needle aspiration biopsy (FNAB) findings; sCt levels; and the referring doctor’s specialty.

RET mutation analysis Genomic DNA was extracted from peripheral blood leucocytes using an in-house protocol (6). Sequence analysis of hot-spot-bearing exons 8, 10, 11, and 13-16 was performed, and extended RET gene analysis was also conducted (7) in all patients considered to have sporadic disease according to hot-spot exon 8, 10, 11, 13-16 analysis who presented with multifocal MTC or Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

Clinical evaluation and follow-up after RET screening In agreement with their physicians, patients with a diagnosis of MTC and the mutation-carrier relatives were subjected to further biochemical and imaging evaluations. Some of the patients continued to be followed up in the Ceará BRASMEN Center. Patients with MTC who had already undergone surgical treatment at the time of RET genetic screening were initially followed with sCt, CEA, ionized calcium, TSH, and cervical US assessments every 4 months. Imaging procedures to detect metastases, such as abdominal magnetic resonance (MRI) or 3-phase contrast-enhanced multidetector liver CT, thoracic CT, column and sacral MRI and bone scans, were performed whenever sCt levels were higher than 150 pg/mL. Patients with hereditary MTC and their mutation-carrier relatives who had already undergone surgical treatment were followed with the same complementary tests, as well as annual 24-hour urine catecholamine and metanephrine assessments. Patients who were diagnosed with MTC after RET screening were encouraged to undergo specific surgical treatments according to their preoperative sCt and imaging results, and mutation carriers were encouraged to undergo additional evaluations to screen for PHEO, HPTH, and other MEN 2 syndrome components.

Histopathology The histological diagnosis of MTC was rendered according to the 2015 ATA guidelines (1), according to the presence of the basic histological pattern of the disease (i.e., typically round, polyhedral, or spindleshaped and formed sheets or nests, with peripheral palisading in the vascular stroma, and the presence of stromal amyloid) and the typical immunohistochemical (cells positive for calcitonin, synaptophysin and chromogranin) findings.

STATISTICAL ANALYSIS The results are presented as the mean ± standard deviation (SD) and as ranges (minimum-maximum). MannWhitney U tests and analyses of variance (ANOVA) were used to compare quantitative variables between two and Arch Endocrinol Metab. 2018;62/6

three or more groups, respectively, and Fisher’s exact test was used to analyze changes in categorical variables. SPSS software (version 23; SPSS Inc., Chicago, IL, USA) was used to perform the analysis. Statistical significance was indicated by p < 0.05.

RESULTS Patients and relatives Overall, over the last 5 years, we performed 263 RET sequencing tests in the BRASMEN-Ceará study. Sixteen patients met one or more of the exclusion criteria described above. Therefore, we performed RET genetic screening in 247 subjects, including 60 index patients with MTC and 187 at-risk relatives, from the State of Ceará, Brazil. Head-and-neck surgeons referred most of the index patients (Table 1). During the initial phase of the study, based only on RET-screened patient clinical history and physical examination results, we identified only 6 index patients and three relatives with hereditary MTC. These patients were characterized as hereditary index patients because they had another family member with MTC or a personal/familial history of MEN 2. In the second phase of the study, after performing RET sequencing, we observed that 23 MTC index patients who were initially classified as sporadic disease patients were, in fact, hereditary index patients. Regarding at-risk RET-screened relatives, we found that 73 relatives carried mutations, and 114 relatives were non-carriers (Figure 1 and Table 2). The clinical and genetic data of the MTC index patients and the mutation-carrier relatives are shown in Table 2. The mutation profile of RET mutations found in Ceará are shown in Figure 2. Overall, of the 29 hereditary MTC index patients who underwent RET screening, 28 definitely underwent TT, and the remaining patient was lost to follow-up (Table 2). In all but one of the MTC index patients, RET screening was performed only postoperatively (Table 1). Regarding the mutation-carrier relatives, 25 patients presented MTC/CCH confirmed by either histopathology after TT (n = 24) or by FNAB cytology positive for MTC along with elevated sCt levels above 100 pg/mL (n = 1) (Table 2). Among those 24 patients with positive histopathology, 6 had already been diagnosed with MTC before RET screening, as they underwent evaluations for asymptomatic thyroid nodules because they knew about index cases in their 625

Copyright© AE&M all rights reserved.

were young and experienced an unfavorable follow-up course (7).


RET screening in the Northeast of Brazil

families (2 relatives of index patient #4, 1 relative of index patient #12) or because they noticed thyroid nodule growth but were not aware that they had a

family member with MTC until a broad pedigree was performed (3 relatives of index patient #20, Table 2). In the remaining patients (n = 18), RET screening led

Table 1. Preoperative clinical data and RET screening results for the index patients with sporadic and hereditary disease and RET mutation-carrier relatives with confirmed diagnoses of MTC/CCH after total thyroidectomy Index Clinical data

Copyright© AE&M all rights reserved.

Gender (Male:Female) Age at RET screening (years) Mean ± SD Median (range) RET screening Before TT After TT Age at diagnosis of MTC/CCH (years) Mean ± SD Median (range) Previous knowledge of MTC/CCH diagnosis at TT Not known By LN biopsy By thyroid nodule/LN FNAB cytology and level of sCt > 100 pg/mL Probable (sCt > 100 pg/mL without + cytology for MTC) Characteristics of preoperative USc Without thyroid nodules at US Not available Suspicious thyroid nodule at US Non-suspicious thyroid nodule at US Preoperative cytology Not performed/Result not available Insufficient Insular neoplasia Benign: paucicellular/satisfactory cellularity Atypia of indeterminate significance Follicular Suggestive of PTC/PTC Suggestive of MTC/MTC Suggestive of anaplastic carcinoma Preoperative sCt (pg/mL)d Mean ± SD Median (range) Reference for RET screening Endocrinologist Head and Neck surgeon General surgeon Oncologist Family member

Relativesb Before RET After RET screening (n = 6) screening (n = 18) 1:5 7:11 n = 6 (100%) n = 18 (100%) 52 ± 23 36 ± 21 49 (29 – 93) 32 (7 – 77) n = 6 (100%) n = 18 (100%) 6 0 0 18 n = 6 (100%) n = 18 (100%) 42 ± 17 37 ± 21 48 (17 – 57) 33 (8 – 77) n = 6 (100%) n = 18 (100%) 6 9 0 0

Sporadic (n = 31) 7:24 n = 31 (100%) 50 ± 13 50 (22 – 37) n = 31 (100%) 1 30 n = 31 (100%) 48 ± 15 50 (20 – 73) n = 31 (100%) 18 2

Hereditary (n = 28)a 7:21 n = 28 (100%) 48 ± 15 52 (16 – 71) n = 28 (100%) 0 28 n = 28 (100%) 43 ± 14 42 (16 – 65) n = 28 (100%) 22 1

10

5

0

2

1 n = 31 (100%) 0 12 14 5 n = 24 (77%) 6/1 0 1 1/6 1 4 0 10 1 n = 8 (26%) 855 ± 1389 159 (19-4020) n = 31 (100%) 9 22 0 0 0

0 n = 28 (100%) 0 12 10 6 n = 17 (61%) 8/4 1 0 2/2 0 4 3 5 0 n = 5 (18%) 1266 ± 1929 347 (12-4640) n = 28 (100%) 5 21 1 1 0

0 n = 6 (100%) 0 2 4 0 n = 4 (67%) 1/1 0 0 2/1 0 1 0 0 0 n=0 n = 6 (100%) 1 3 0 0 2

7 n = 18 (100%) 1 0 10 7 n = 11 (61%) 7/0 0 0 0/2 5 0 2 2 0 n = 17 (94%) 343 ± 709 98 (8-2695) n = 18 (100%) 0 1 0 0 17

p < 0.05e NS

NS

< 0.05f

NS

NS

NS

MTC: medullary thyroid cancer; CCH: C-cell hyperplasia; SD: standard deviation; TT: total thyroidectomy; LN: lymph node; FNAB: fine-needle aspiration biopsy; sCt: serum calcitonin; US: ultrasound; PTC: papillary thyroid cancer. a One index patient was lost to follow-up before TT. b One mutation-carrier relative did not undergo TT. c Nodules were considered suspicious if they exhibited one of the following characteristics: size greater than 3 cm, microcalcifications, or central flow on Doppler US, and suspected lymph node. d Cut-off values of 18.4 pg/mL for males and 7.8 pg/mL for females. e Fisher’s exact test. f Not known versus known (Fisher’s exact test)

626

Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

to the diagnosis of MTC/CCH (Tables 1 and 2). In 2 of these 18 patients, we found only CCH. Only one mutation-carrier relative referred to TT had neither

MTC nor CCH on histopathology. She was a p.V804L mutation carrier and had undergone TT at 9 years of age.

Table 2. Genetic and clinical features of the 29 patients considered index cases upon RET sequencing

Index cases

Age at RET sequencing (years)

Age at MTC diagnosis (years)

Relatives affected by MTC at time of screening

1b

58

59

0

2b

62

30

0

3b

35

34

Months of follow up

Mutation carriers/ screened relatives

MTC

10

MTC

36

p.C630R

MTC

RET mutation

Phenotype

10

p.C609G

10

p.C611R

0

10

Exon

Relatives carrying mutation with MTC/ CCHa Before RET screening

After RET screening

0/2

0

0

0

0

0

29

1/6

0

1

4

26

22

3

11

p.C634R

MTC + PHEO + CLA

47

6/13

2

3

5b

22

21

0

11

p.C634R

MTC

14

1/2

0

0

6

43

37

0

11

p.C634R

MTC + PHEO

2

0

0

0

7

40

20

0

11

p.C634R

MTC + PHEO + CLA

42

1/4

0

1

8b

41

41

0

13

p.L790F

MTC

16

0

0

0

9b

63

61

0

14

p.V804L

MTC

21

5/10

0

2

b

10

51

39

0

14

p.V804L

MTC

10

5/10

0

0

11b

54

54

0

14

p.V804L

MTC

3

0

0

0

12

67

56

1

14

p.V804L

MTC

4

1/1

1

0

b

13

42

42

0

14

p.V804L

MTC

69

1/3

0

0

14b

52

49

0

14

p.V804L

MTC

59

3/6

0

2

b

15

71

65

0

14

p.V804L

MTC

8

0/1

0

0

16b

61

55

0

14

p.V804M

MTC

8

1/1

0

1

17

45

41

1

14

p.V804M

MTC

3

2/3

0

0

b

18

43

43

0

14

p.V804M

MTC

5

2/2

0

0

19b

36

36

0

15

p.S891A

MTC

76

1/4

0

0

20b

54

54

0

16

p.M918V

MTC

67

33/95

3

7

b

21

59

58

0

16

p.M918V

MTC

21

0/0

0

0

22b

55

55

0

16

p.M918V

MTC

70

1/5

0

0

23b

42

40

0

16

p.M918V

MTC

70

4/7

0

1

b

24

66

59

0

16

p.M918V

MTC

71

3/3

0

0

25b

28

24

0

16

p.M918V

MTC

74

1/3

0

1

26b

55

55

0

16

p.M918V

MTC

51

0/0

0

0

b

27

54

40

0

16

p.M918V

MTC

52

0/0

0

0

28b

69

58

0

16

p.M918V

MTC

15

1/4

0

0

29

16

16

0

16

p.M918T

MEN 2B

1

0/2

0

0

TOTAL (mean ± SD)

48.6 ± 14.4

43.6 ± 14.1

5

--

--

--

32.9 ± 27.3

73/187 (39%)

6

19

Median (range)

52 (16-71)

42 (16-65)

--

--

--

--

21 (1-76)

--

--

--

c

hyperparathyroidism; SD: standard deviation. This column refers only to relatives who underwent RET screening. Relatives who probably carry RET mutations (by clinical assessment only) but did not undergo RET screening were not included. Diagnoses of MTC/CCH were made based on histopathology results after TT (n = 18) or when relatives who had not undergone TT had fine-needle aspiration cytology positive for MTC concurrently with elevated serum calcitonin levels (above 100 pg/mL) (n = 1). b Patients with apparent sporadic disease based on their clinical assessment and RET screening results. c Not significant (Mann-Whitney U test).

a

Arch Endocrinol Metab. 2018;62/6

627

Copyright© AE&M all rights reserved.

RET: REarranged during Transfection protooncogene; MTC: medullary thyroid cancer; CCH: C-cell hyperplasia; PHEO: pheochromocytoma; CLA: cutaneous lichen amyloidosis; HPTH: primary


RET screening in the Northeast of Brazil

Genotype-phenotype correlation

Subjects with inclusion criteria n = 247

At first clinical assessment

Sporadic index n = 54

Hereditary indexa n=6

After RET screeening

Sporadic index n = 31

Hereditary index n = 29

CopyrightŠ AE&M all rights reserved.

628

Relatives without mutation n = 114

30 25 20 15 10 5 8T 91

8V 91

p.M

1A 89

p.M

4M

p.S

4L 80

80 p.V

79

0F

p.V

4R

p.L

63

0R

p.C

63

9G 60

p.C

60

9G

0 p.C

The preoperative data of the RET-screened patients who underwent TT are shown in Table 1. The majority of patients were women. Additionally, the mutationcarrier relatives whose neoplasm diagnoses were made after RET screening were younger than the patients whose neoplasm diagnoses were made before genetic analysis (not significant). In most patients, MTC/CCH was diagnosed based on histopathological findings following TT because only 20% of patients had cytology results suggestive of MTC (Figure 3), and consequently, only a few patients had preoperative sCt measurements. However, as expected, we observed lower levels of sCt in mutation-carrier relatives than in hereditary index patients, although the difference in tumor marker levels between the two groups was not statistically significant (Table 1). In 9 of the 18 mutation-carrier relatives diagnosed with MTC/CCH after RET screening, diagnoses for MTC had already been made possible by their clinical evaluations, as they had sCt levels above 100 pg/mL along with a suspicious thyroid nodule on US and/or FNAB thyroid nodule or cervical LN cytology results that were suspicious for malignancy (Table 1).

Mutation carriers relatives n = 73

35

%

Preoperative evaluation of RET-screened patients

At-risk relatives n = 187 Probably mutation carriersa n = 3/187

Figure 1. Fluxogram of the study according to the first clinical assessment or RET screening. aProbable RET mutation carriers identified before genetic screening due to either a personal history of MEN 2 or physical examination findings suggestive of MEN 2.

p.C

Based on the clinical evaluation and RET genetic screening results, we identified 29 index patients with hereditary MTC and 31 index patients with sporadic MTC (Figure 1). The most frequent mutations found were p.V804L and p.M918V (Table 2, Figure 2). PHEO was associated with the following mutations: p.C634R, p.V804L, and p.M918T. HPTH was associated with p.C634R and p.V804L, and LCA was associated with p.C634R. There were no cases of MEN 2A with HD (Table 2). Patients with the p.M918B mutation did not present the MEN 2B phenotype (8). We identified only one patient with MEN 2B, who had the classical p.M918V mutation. Unsuspected germline RET mutations were found in 23 of the 54 MTC patients (43%) who presented with apparent sporadic disease, according to their negative familial histories and MTC-only phenotypes. These patients were informed about the hereditary origins of their neoplasms, and RET genetic screening was subsequently offered to their relatives (Figure 1).

Subjects with exclusion criteria n = 16

Screened subjects n = 263

Figure 2. Profiles of RET gene mutations in CearĂĄ. Preoperative cytology Suggestive of MTC 20%

Not performed 26%

Suggestive of non MTC 9% Not available 7% Follicular 11% Atypia of indeterminate significance 7%

Insufficient 1% Benign: paucicellular satisfactory cellularity 19%

Figure 3. Preoperative cytology of index patients with sporadic and hereditary disease and their RET mutation-carrier relatives. Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

Postoperative evaluation The postoperative data for the RET-screened patients who underwent TT and received a confirmed diagnosis of MTC are shown in Tables 3 and 4. An interesting finding was that relatives whose diagnoses of MTC/CCH were made after RET screening had lower sCt levels and less frequent postsurgical

hypoparathyroidism than index patients and mutationcarrier relatives whose diagnoses of MTC/CCH were made before RET screening (p < 0.05) (Table 3). Also, up to the most current follow-up, all relatives diagnosed with MTC/CCH after RET screening were alive, while 5 index cases had died (Figure 4, Table 3).

Table 3. Postoperative clinical data for the index patients with sporadic and hereditary disease and RET mutation-carrier relatives with confirmed MTC/ CCH after total thyroidectomy Index Clinical data

Age at TT (years) Mean ± SD Median (range) Clinical presentation At diagnosis/at last medical visit

Relatives

Sporadic (n = 31)

Hereditary (n = 28)

Before RET screening (n = 6)

After RET screening (n = 18)

n = 31 (100%)

n = 28 (100%)

n = 6 (100%)

n = 18 (100%)

47 ± 15

43 ± 14

41 ± 16

35 ± 18

48 (20-73)

42 (16-65)

48 (17-57)

33 (8-67)

n = 31 (100%)

n = 28 (100%)

n = 6 (100%)

n = 18 (100%)

22 / 21

14 / 12

3/0

13 / 13

p

NS

a

Central LN metastases

0/0

5/0

1/0

2/2

Lateral LN metastases

8/4

7 / 12

2/5

3/3

Systemic metastases EBRT

1/6

2/4

0/1

0/0

n = 31 (100%)

n = 28 (100%)

n = 6 (100%)

n=0

Performed

6

7

1

0

Not performed

25

21

5

0

n = 30 (97%)

n = 28 (100%)

n = 6 (100%)

n = 18 (100%)

Yes

7

14

0

1

No

23

14

6

17

Last serum Ct of follow up (pg/mL)

n = 27 (87%)

n = 22 (79%)

n = 6 (100%)

17 (94%)

Mean ± SD

437 ± 1223

324 ± 890

192 ± 317

26 ± 71

Post-surgical hypoparathyroidismb

Median (range) Current follow up

2 (2-5660)

7 (2-3860)

43 (8-818)

2 (2-273)

n = 26 (94%)

n = 24 (86%)

n = 6 (100%)

n = 17 (94%)

At Brasmen-Ceará Center Only

4

3

0

5

At Brasmen-Ceará and original doctor

10

14

5

8

At original doctor only

9

7

1

4

Lost of follow up

5

2

0

1

Died

3

2

0

0

n = 31 (100%)

n = 28 (100%)

n = 6 (100%)

n = 18 (100%)

19 ± 22

35 ± 27

36 ± 18

40 ± 17

8 (2-76)

34 (1-76)

44 (2-47)

41 (3-67)

n = 1 (3%)

0

0

0

Time of follow-up (months) Mean ± SD Median (range) ITK use

NS

NS

< 0.05c

< 0.05d

NS

CCH: C-cell hyperplasia; EBRT: external beam radiation therapy; FNAB: fine-needle aspiration biopsy; LN: lymph node; ITK: tyrosine kinase inhibitor; MTC: medullary thyroid cancer; PTC: papillary thyroid cancer; RET: REarranged during Transfection protooncogene; sCt: serum calcitonin; SD: standard deviation; TT: total thyroidectomy; NS: not significant. It was considered the most severe clinical presentation (e.g., when patients had metastatic LNs and systemic metastases, it was allocated only into systemic metastases. In some patients, the diagnosis of MTC was discovered many years after TT). a

Not possible to determine in one patient with sporadic MTC because he underwent total thyroidectomy together with total parathyroidectomy for the treatment of hyperparathyroidism caused by parathyroid hyperplasia related to MEN 1 syndrome. Thyroidectomy was performed because of intraoperative suspicion for parathyroid carcinoma. Thyroid tissue histopathology revealed the presence of a microscopic MTC. b

c

Fisher’s exact test among all groups.

d

ANOVA test among index patients with hereditary disease, relatives before RET screening, and relatives after RET screening.

Arch Endocrinol Metab. 2018;62/6

629

Copyright© AE&M all rights reserved.

Without LN and/or systemic metastases


RET screening in the Northeast of Brazil

Regarding pathologic tumor and LN staging, we observed that relatives whose diagnoses of MTC/CCH were made after RET screening had smaller tumor dimensions than index patients (p < 0.05), although there was no significant difference in the frequency of LN involvement between the two groups (Figure 5, Table 4).

A higher proportion of index patients with hereditary disease and their relatives presented tumor multifocality than did index patients with sporadic disease, although the percentage of multifocality was not negligible (22%) in the latter group (Table 4). This finding led us to perform extended RET gene analysis

Table 4. Postoperative staging of the index patients with sporadic and hereditary disease and RET mutation-carrier relatives with histopathologically confirmed MTC/CCH after total thyroidectomy Index Sporadic (n = 31)

Hereditary (n = 28)

Before RET screening (n = 6)

After RET screening (n = 18)

n = 31 (100%)

n = 28 (100%)

n = 6 (100%)

n = 18 (100%)

CCH only

0

0

0

2

pTx

2

3

1

1

pT1a

12

7

0

9

pT1b

2

4

0

5

pT2

6

4

2

1

pT3

6

8

3

0

pT4a

2

2

0

0

pT4b

1

0

0

0

pNx

13

14

3

4

pN0

10

2

0

10

pN1a

0

5

2

1

pN1b

8

7

1

3

n = 27 (87%)

n = 27 (96%)

n = 5 (83%)

n = 15 (94%)

6

22

4

12c

21

5

1

3

2/ 2

1/ 0

1/0

1/0

n = 16 (52%)

n = 27 (96%)

n = 5 (83%)

n = 17 (94%)

Yes

12

18

4

7d

No

4

9

1

10

Not performed

15

1

1

1

n = 29 (94%)

n = 27 (96%)

n = 5 (83%)

n = 17 (94%)

3

3

1

2d

26

24

4

15

2

1

1

1

Pathology data

Pathologic tumoral and lymph node staging

Multifocality Yes No Unknown / Not possible due to large mass a

IHC positive for MTC/CCH

Association with PTC Yes No CopyrightŠ AE&M all rights reserved.

Relatives

Unknown

a

b

pc

< 0.05

0.03

< 0.05

NS

NS

Not possible due to previous partial thyroidectomy and unavailable histopathology results. Assessment of multifocality was also not possible in large tumors occupying the entire thyroid. During the initial stage of the disease, it was not possible to determine if there were multiple tumor foci that coalesced or if there was a unique focus that increased in size with time. c Chi-square test. RET: REarranged during Transfection protooncogene; CCH: C-cell hyperplasia; IHC: immunohistochemistry; MTC: medullary thyroid cancer; PTC: papillary thyroid cancer; NS: not significant. a

b

630

Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

30

previously undergone a partial thyroidectomy and did not have available histopathology results.

20

DISCUSSION

15 10 5 0

Sporadic index

Hereditary index Alive

Relatives before RET

Lost of follow up

Relatives after RET Died

Figure 4. Current follow-up status of index (sporadic and hereditary) patients and RET-carriers relatives who underwent total thyroidectomy.

Tumor Staging

A

p = 0.4 Fisher test

100 80 % 60 40 20 0

Index T1/T2

p = 0.4 Fisher test

after RET

Relatives after RET T3/T4

Lymph node (LN) involvement

B 100

p = 0.5 Fisher test

80 % 60 40 20 0

Index

Relatives after RET LN -

LN +

Figure 5. Postoperative staging of the index patients with sporadic and hereditary disease and RET mutation-carrier relatives.

in index patients with sporadic disease to exclude the presence of mutations in other exons. However, no patients presented with new mutations in extended RET gene analysis. We observed simultaneous MTC and papillary thyroid carcinoma (PTC) in 9 of 78 (11.5%) patients with MTC (Table 4). In some patients, this association could not be evaluated because these patients had Arch Endocrinol Metab. 2018;62/6

Identification of RET mutations in patients with a diagnosis of MTC is essential, both for index patients and for at-risk relatives, and allows for the potential identification of hereditary MTC patients appearing to have sporadic MTC who are not followed-up regarding other MEN 2A components, especially the lifethreatening disease PHEO (1). For patients who are already clinically suspected of having hereditary MTC, this analysis allows identification of the specific RET mutation and an estimation of the disease’s likely clinical course. For affected family members, RET mutation analysis allows MTC to be cured when prophylactic thyroidectomy is performed and also allows precocious Lymph node (LN) involvement p = 0.5 B detection of and administration of therapy Fisher test for other 100 components of the syndrome. For non-affected family 80 members, it spares these patients from having to % 60undergo clinical and biochemical monitoring for all 40MEN 2A components (9). Using clinical history and physical examination 20 findings alone, we identified only 6 hereditary MTC 0 Index after RET index cases. However, RETRelatives screening allowed us to LN LN + identify unsuspected germline RET mutations in 23 of the 54 MTC patients (43%) who appeared to present as sporadic disease cases, according to their negative family histories and isolated MTC phenotype (Table 2). Therefore, through RET screening, the number of index patients with hereditary MTC increased to 29. Elisei and cols. also found unsuspected germline RET mutations in some sporadic cases but at a percentage (only 7.3% of cases) that was much lower than in our study (5). We believe that some particular findings of this study may explain this difference, such as the higher prevalence of the p.M918V mutation (> 30%) among hereditary MTC cases (Figure 2) marked by the isolated MTC phenotype, as well as the higher percentage of hereditary index patients (48%) in this study compared to the 25% usually described in the literature (5,10-12). Regarding this higher percentage of hereditary index patients in Ceará compared to the literature, we believe that this may reflect the high frequency of mutation dispersion in the past, before the existence of birth control policies. Families were larger and comprised many children. In addition, the frequency of extramarital relationships, which has been 631

Copyright© AE&M all rights reserved.

Number of patients

25


Copyright© AE&M all rights reserved.

RET screening in the Northeast of Brazil

reported to be a prevalent practice, was higher and may have resulted in an increased number of births outside of marriage (13). In our initial evaluation, the most common mutation found in Ceará was p.M918V (Figure 2). Thus far, this mutation has been identified in 9 families who were apparently unrelated. Previously, this mutation had been described in only one patient with MTC and in 1 family member without a neoplasm in the literature (14). As the number of index patients who were found to have the p.M918V mutation was increased, we observed that some of those patients lived in nearby regions, such as in the Northeastern region of Ceará State. This finding led us to question whether a founder effect could explain the high frequency of the p.M918V mutation in Ceará, as at that time, 8 families with a p.M918V mutation had already been identified. The evidence suggestive of a founder effect as an explanation for the frequency of the p.M918V mutation in Ceará led us to consider whether the first 8 families diagnosed with a p.M918V mutation were actually one large family (8). The most frequent mutations identified in our study were in codons 918 and 804 (Figure 2, Table 2), findings that differ from those of other studies worldwide, in which mutations in codon 634 were the most common mutations found (15-27). Also, mutations in codons 338, 515, 666, 618, 620, 768, 848, 883 and 904, which were observed in the European studies (25), have not been identified in Ceará thus far (Table 5). Regarding preoperative evaluations, the low sensitivity of FNAB cytology noted in this study (20%) (Figure 3) had previously been reported in other studies (28-30). Essig Jr and cols. concluded that cytological evaluations alone limited one’s ability to perform an optimal preoperative assessment and initial surgery in over half of affected patients in sporadic MTC (28). The present study led us to analyze the potential benefits of diagnosing affected relatives. Comparison of mutation-carrier relatives diagnosed with MTC after RET screening to index patients showed that those relatives had a lower frequency of complications, such as post-surgical hypoparathyroidism, and lower levels of sCt upon their last visit (p < 0.05). Also, none of the affected relatives died, while 5 indexes succumbed to disease (Figure 4). Interestingly, the affected relatives were not diagnosed at a significantly younger age (Table 3), nor were they diagnosed at a more favorable clinical or pathologic stage than their counterparts (Table 4). In fact, a trend toward diagnosis at a younger age was 632

observed, but the trend was not statistically significant (Table 3). We also observed that relatives who had a diagnosis of MTC after RET screening had smaller tumor dimensions than index patients, but the degree of lymph node involvement was similar between the two groups (Figure 5). In fact, 50% of those mutationcarrier relatives diagnosed with MTC after RET screening already presented clinical evidence of MTC before the RET screening results were available (Table 1). Therefore, in our study population, the benefits of RET screening were not fully realized. We consider that the major reason for this was because these patients received this exam relatively late, as they were not aware of its existence and benefits. We believe that if we can offer earlier RET screening for at-risk patients, we will be able to diagnose neoplasms at an earlier stage, or, more desirably, before they have developed. The finding of simultaneous MTC and PTC in this study (11.5%) had previously been described in previous studies and was observed in the following variable percentages of patients in those studies: 3.6% of patients in Germany (31), 13.8% of patients in Italy (32) and 19% of patients in Korea (33). Those discrepancies were attributed to environmental conditions and differences in the populations of the studies in question (1). Regarding tumor multifocality, this phenomenon was observed more frequently in index patients with hereditary disease and their relatives than in index patients with sporadic disease. Consistent with the findings of the study by Lindsey and cols. (7), in the present study, we noted no direct clinical benefit to extending the RET germline analysis beyond the hot-spot regions in patients with sporadic MTC with multifocality (22%) since the analysis did not identify any mutations in other exons. Therefore, these cases were true sporadic cases of MTC with multifocal tumors, which had been observed in a recent study (34). Our study population comprised a significant number of mutation carriers who did not attend follow-up visits after learning their RET sequencing results, nor did they bring their at-risk relatives to a clinic to undergo RET screening. Geographic distances and some patients’ poor financial statuses, which prevented them from bearing the financial burden imposed by traveling to our center, are recognized as important factors that delayed the performance of RET screening. We also considered other reasons that may explain some patients’ delays in undergoing genetic analysis and some patients’ refusal to undergo Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

Table 5. Prevalence of RET gene mutations in the State of Ceará along with major international studies and their respective genotype-phenotype correlations

RET mutations

Exon

ATA risk

MEN 2A

MEN 2B

Ceará, Brazil

ITAMEN (22)

Germany, Halle 19942012 (25)

EUROMEN (23)

France, multicentric (21, 23)

n (%)

n (%)

n (%)

n (%)

n (%)

p.T338I

5

MOD

X

0

1 (0.4)

0

0

0

p.C515S

8

MOD

X

0

1 (0.4)

0

0

0

p.C609F/G/G/R/S/Y

10

MOD

X

1 (3)

6 (2.5)

1 (0.5)

1 (0.7)

1 (1)

p.C611F/G /R/S/W/Y

10

MOD

X

1 (3)

1 (0.4)

6 (3.1)

4 (2.8)

1 (1)

p.C618G/R/R//F/S/Y

10

MOD

X

0

15 (6.1)

11 (5.8)

10 (7)

6 (6)

p.C620 F/F/G/R/S/W/Y

10

MOD

X

0

9 (3.7)

14 (7.3)

10 (7)

12 (12)

p.C630 F/R/R/S/Y

11

MOD

X

1 (3)

4 (1.6)

1 (0.5)

1 (0.7)

0

p.C634F/G/R/S/W/Y

11

HIGH

X

4 (14)

86 (35.2)

73 (38.2)

98 (69)

46 (47)

p.K666M

11

MOD

X

0

1 (0.4)

0

0

0

p.E768D

13

MOD

X

0

9 (3.7)

2 (1)

1 (0.7)

2 (2)

p.L790F

13

MOD

X

1 (3)

8 (3.3)

26 (13.6)

7 (4.9)

4 (4)

p.V804L/M/M

14

MOD

X

10 (34)

52 (21.3)

19 (10)

3 (2.1)

15 (15)

p.M848T

14

MOD

X

0

1 (0.4)

0

0

0

p.A883T

15

MOD

X

0

1 (0.4)

0

0

0

p.S891A/A

15

MOD

X

1 (3)

23 (9.4)

6 (3)

3 (2.1)

7 (7)

p.S904F

14

MOD

X

0

1 (0.4)

0

0

0

p.M918T

16

HIGHEST

1 (3)

17 (7.0)

32 (16.8)

4 (2.8)

3 (3)

p.M918V

16

MOD

X

9 (31)

2 (0.8)

0

0

0

Without mutations

_

_

X

0

6 (2.5)

0

0

0

29 (100)

244

191 (100)

142 (100)

97 (100)

X

TOTAL

genetic analysis. First, not every RET gene carrier is emotionally prepared to face a diagnosis of MTC or to share personal genetic information with his or her relatives. These types of behavior may be related to feelings of guilt and resentment, emotional distress, and poor familial interactions (25,35). Other obstacles affecting patients’ abilities to undergo genetic analysis are related to individual aspects, such as patients’ understanding of their disease and religious beliefs. In contrast to the above findings, in some families, we observed solidarity among family members. Additionally, we noted that some family members felt a need to reveal their diagnosis and their intention to Arch Endocrinol Metab. 2018;62/6

face their disease positively in order to motivate their relatives to accelerate their diagnostic and treatment processes to avoid being diagnosed with a neoplasm at the same stage that they had been. In conclusion, in our study, RET screening was profoundly crucial for the early diagnosis of MTC and facilitated monitoring of other MEN 2A components and identification of unsuspected germline mutations in index patients with apparent sporadic disease. We noted a mutation profile that differed from that of previous European studies. We attribute these differences to our having restricted our analysis to the State of Ceará and to our having evaluated a small number of 633

Copyright© AE&M all rights reserved.

The mutations causing MEN 2A and MEN 2B are shown in black X, whereas the FMTC mutations are shown in bold X. ATA: American Thyroid Association; MTC: medullary thyroid carcinoma; FMTC: familial medullary thyroid carcinoma; MOD: moderate; RET: REarranged during Transfection; MEN: multiple endocrine neoplasia. Modified from references 4 and 25.


RET screening in the Northeast of Brazil

patients. These features make the extrapolation of our results to the rest of the country unlikely, as different colonization patterns occurred throughout Brazil. However, we believe that any regional differences in RET distributions that exist are likely to diminish as a result of dynamic world social, political and economic scenarios impacting migration processes. Acknowledgments: we would like to express our sincere appreciation to the medical team of Ceará (including the endocrinologists, head-and-neck surgeons, general surgeons, oncologists, nuclear medicine physicians, radiotherapists, radiologists, and pathologists), whose cooperation made this work possible. We would like to give our posthumous thanks to Mr. Antônio Adriano Mendes, who kindly collaborated with us to produce the pedigrees. We also thank Ângela Maria Faria for her administrative assistance and the Federal Agency for the Support and Evaluation of Graduate Education (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – CAPES), the National Council for Scientific and Technological Development (CNPq) and the São Paulo Research Foundation (Fundação de Amparo à Pesquisa do Estado de São Paulo – Fapesp) for providing financial support. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1.

Wells SA Jr., Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, et al. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid. 2015;25:567-610.

2. Instituto Nacional de Câncer José Alencar Gomes da Silva. Estimativa 2016: Incidência de Câncer no Brasil. CdPeV, editor. Rio de Janeiro: INCA; 2015. 122 p. 3. Veiga LH, Neta G, Aschebrook-Kilfoy B, Ron E, Devesa SS. Thyroid cancer incidence patterns in Sao Paulo, Brazil, and the U.S. SEER program, 1997-2008. Thyroid. 2013;23:748-57. 4. Wells SA Jr, Pacini F, Robinson BG, Santoro M. Multiple endocrine neoplasia type 2 and familial medullary thyroid carcinoma: an update. J Clin Endocrinol Metab. 2013;98:3149-64. 5. Elisei R, Romei C, Cosci B, Agate L, Bottici V, Molinaro E, et al. RET genetic screening in patients with medullary thyroid cancer and their relatives: experience with 807 individuals at one center. J Clin Endocrinol Metab. 2007;92:4725-9. 6. Kizys MM, Cardoso MG, Lindsey SC, Harada MY, Soares FA, Melo MC, et al. Optimizing nucleic acid extraction from thyroid fineneedle aspiration cells in stained slides, formalin-fixed/paraffinembedded tissues, and long-term stored blood samples. Arq Bras Endocrinol Metabol. 2012;56:618-26.

Copyright© AE&M all rights reserved.

7.

Lindsey SC, Kunii IS, Germano-Neto F, Sittoni MY, Camacho CP, Valente FO, et al. Extended RET gene analysis in patients with apparently sporadic medullary thyroid cancer: clinical benefits and cost. Horm Cancer. 2012;3:181-6.

8. Martins-Costa MC, Cunha LL, Lindsey SC, Camacho CP, Dotto RP, Furuzawa GK, et al. M918V RET mutation causes familial medullary thyroid carcinoma: study of 8 affected kindreds. Endocr Relat Cancer. 2016;23:909-20.

634

9. Frank-Raue K, Raue F. Hereditary Medullary Thyroid Cancer Genotype-Phenotype Correlation. Recent Results Cancer Res. 2015;204:139-56. 10. Maia AL, Siqueira DR, Kulcsar MA, Tincani AJ, Mazeto GM, Maciel LM. Diagnosis, treatment, and follow-up of medullary thyroid carcinoma: recommendations by the Thyroid Department of the Brazilian Society of Endocrinology and Metabolism. Arq Bras Endocrinol Metabol. 2014;58:667-700. 11. Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib H, et al. Medullary thyroid cancer: management guidelines of the American Thyroid Association. Thyroid. 2009;19:565-612. 12. Pacini F, Castagna MG, Brilli L, Pentheroudakis G. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21 Suppl 5:v214-9. 13. Freyre G. Casa Grande e Senzala. Edição comemorativa 80 anos ed. Rio de Janeiro: Global; 2012. 728 p. 14. Cosci B, Vivaldi A, Romei C, Gemignani F, Landi S, Ciampi R, et al. In silico and in vitro analysis of rare germline allelic variants of RET oncogene associated with medullary thyroid cancer. Endocr Relat Cancer. 2011;18:603-12. 15. Sanso GE, Domene HM, Garcia R, Pusiol E, de M, Roque M, et al. Very early detection of RET proto-oncogene mutation is crucial for preventive thyroidectomy in multiple endocrine neoplasia type 2 children: presence of C-cell malignant disease in asymptomatic carriers. Cancer. 2002;94:323-30. 16. Alvandi E, Akrami SM, Chiani M, Hedayati M, Nayer BN, Tehrani MR, et al. Molecular analysis of the RET proto-oncogene key exons in patients with medullary thyroid carcinoma: a comprehensive study of the Iranian population. Thyroid. 2011;21:373-82. 17. Sharma BP, Saranath D. RET gene mutations and polymorphisms in medullary thyroid carcinomas in Indian patients. J Biosci. 2011;36:603-11. 18. Chung YJ, Kim HH, Kim HJ, Min YK, Lee MS, Lee MK, et al. RET proto-oncogene mutations are restricted to codon 634 and 618 in Korean families with multiple endocrine neoplasia 2A. Thyroid. 2004;14:813-8. 19. Zhou Y, Zhao Y, Cui B, Gu L, Zhu S, Li J, et al. RET proto-oncogene mutations are restricted to codons 634 and 918 in mainland Chinese families with MEN2A and MEN2B. Clin Endocrinol (Oxf). 2007;67:570-6. 20. Qi XP, Chen XL, Ma JM, Du ZF, Fei J, Yang CP, et al. RET protooncogene genetic screening of families with multiple endocrine neoplasia type 2 optimizes diagnostic and clinical management in China. Thyroid. 2012;22:1257-65. 21. Niccoli-Sire P, Murat A, Rohmer V, Franc S, Chabrier G, Baldet L, et al. Familial medullary thyroid carcinoma with noncysteine ret mutations: phenotype-genotype relationship in a large series of patients. J Clin Endocrinol Metab. 2001;86:3746-53. 22. Romei C, Mariotti S, Fugazzola L, Taccaliti A, Pacini F, Opocher G, et al. Multiple endocrine neoplasia type 2 syndromes (MEN 2): results from the ItaMEN network analysis on the prevalence of different genotypes and phenotypes. Eur J Endocrinol. 2010;163:301-8. 23. Nguyen L, Niccoli-Sire P, Caron P, Bastie D, Maes B, Chabrier G, et al. Pheochromocytoma in multiple endocrine neoplasia type 2: a prospective study. Eur J Endocrinol. 2001;144:37-44. 24. Machens A, Niccoli-Sire P, Hoegel J, Frank-Raue K, van Vroonhoven TJ, Roeher HD, et al. Early malignant progression of hereditary medullary thyroid cancer. N Engl J Med. 2003;349:1517-25. 25. Machens A, Lorenz K, Sekulla C, Hoppner W, Frank-Raue K, Raue F, et al. Molecular epidemiology of multiple endocrine neoplasia 2: implications for RET screening in the new millenium. Eur J Endocrinol. 2013;168:307-14. Arch Endocrinol Metab. 2018;62/6


RET screening in the Northeast of Brazil

26. Toledo RA, Hatakana R, Lourenco DM Jr., Lindsey SC, Camacho CP, Almeida M, et al. Comprehensive assessment of the disputed RET Y791F variant shows no association with medullary thyroid carcinoma susceptibility. Endocr Relat Cancer. 2015;22:65-76. 27. Toledo RA, Maciel RM, Erlic Z, Lourenco DM Jr., Cerutti JM, Eng C, et al. RET Y791F Variant Does Not Increase the Risk for Medullary Thyroid Carcinoma. Thyroid. 2015;25:973-4. 28. Essig GF Jr, Porter K, Schneider D, Debora A, Lindsey SC, Busonero G, et al. Fine needle aspiration and medullary thyroid carcinoma: the risk of inadequate preoperative evaluation and initial surgery when relying upon FNAB cytology alone. Endocr Pract. 2013;19:920-7. 29. Bugalho MJ, Santos JR, Sobrinho L. Preoperative diagnosis of medullary thyroid carcinoma: fine needle aspiration cytology as compared with serum calcitonin measurement. J Surg Oncol. 2005;91:56-60.

31. Machens A, Dralle H. Simultaneous medullary and papillary thyroid cancer: a novel entity? Ann Surg Oncol. 2012;19:37-44. 32. Biscolla RP, Ugolini C, Sculli M, Bottici V, Castagna MG, Romei C, et al. Medullary and papillary tumors are frequently associated in the same thyroid gland without evidence of reciprocal influence in their biologic behavior. Thyroid. 2004;14:946-52. 33. Kim WG, Gong G, Kim EY, Kim TY, Hong SJ, Kim WB, et al. Concurrent occurrence of medullary thyroid carcinoma and papillary thyroid carcinoma in the same thyroid should be considered as coincidental. Clin Endocrinol (Oxf). 2010;72:256-63. 34. Essig GF Jr, Porter K, Schneider D, Arpaia D, Lindsey SC, Busonero G, et al. Multifocality in Sporadic Medullary Thyroid Carcinoma: An International Multicenter Study. Thyroid. 2016;26:1563-72. 35. Machens A, Dralle H. Multiple endocrine neoplasia type 2: achievements and current challenges. Clinics (Sao Paulo). 2012;67 Suppl 1:113-8.

CopyrightŠ AE&M all rights reserved.

30. Chang TC, Wu SL, Hsiao YL. Medullary thyroid carcinoma: pitfalls in diagnosis by fine needle aspiration cytology and relationship

of cytomorphology to RET proto-oncogene mutations. Acta Cytol. 2005;49:477-82.

Arch Endocrinol Metab. 2018;62/6

635


original article

Rare complications of multikinase inhibitor treatment Fabián Pitoia1, Angélica Schmidt1, Fernanda Bueno1, Erika Abelleira1, Fernando Jerkovich1

ABSTRACT Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina 1

Correspondence to: Fabián Pitoia fpitoia@intramed.net Received on Mar/18/2018 Accepted on Oct/30/2018 DOI: 10.20945/2359-3997000000090:

Objective: The advent of multikinase inhibitor (MKI) therapy has led to a radical change in the treatment of patients with advanced thyroid carcinoma. The aim of this manuscript is to communicate rare adverse events that occurred in less than 5% of patients in clinical trials in a subset of patients treated in our hospital. Subjects and methods: Out of 760 patients with thyroid cancer followed up with in our Division of Endocrinology, 29 (3.8%) received treatment with MKIs. The median age at diagnosis of these patients was 53 years (range 20-70), and 75.9% of them were women. Sorafenib was prescribed as first-line treatment to 23 patients with differentiated thyroid cancer and as second-line treatment to one patient with advanced medullary thyroid cancer (MTC). Vandetanib was indicated as first-line treatment in 6 patients with MTC and lenvatinib as second-line treatment in two patients with progressive disease under sorafenib treatment. Results: During the follow-up of treatment (mean 13.7 ± 7 months, median 12 months, range 6-32), 5/29 (17.2%) patients presented rare adverse events. These rare adverse effects were: heart failure, thrombocytopenia, and squamous cell carcinoma during sorafenib therapy and squamous cell carcinoma and oophoritis with intestinal perforation during vandetanib treatment. Conclusions: About 3 to 5 years after the approval of MKI therapy, we learned that MKIs usually lead to adverse effects in the majority of patients. Although most of them are manageable, we still need to be aware of potentially serious and rare or unreported adverse effects that can be life-threatening. Arch Endocrinol Metab. 2018;62(6):636-40 Keywords Multikinase inhibitors; rare adverse events; sorafenib; vandetanib; thyroid carcinoma

INTRODUCTION

Copyright© AE&M all rights reserved.

T

he advent of multikinase inhibitor (MKI) therapy has led to a radical change in the treatment of patients with advanced thyroid carcinoma. The use of these drugs should be considered in patients with radioiodine refractory differentiated thyroid cancer (DTC) or medullary thyroid cancer (MTC) with locally progressive or distant metastatic disease, and/ or symptomatic disease that cannot be managed with surgery or local approaches (1). The profile of adverse events of these drugs is usually well known. They were reported in up to 90% of patients in the ZETA, DECISION, and SELECT studies (2-4). The adverse events that occurred in less than 5% of patients in the clinical trials were considered rare. In the ZETA study (2), acute heart failure, aspiration pneumonia, respiratory arrest, respiratory failure, and staphylococcal sepsis were described in patients on the vandetanib arm with a frequency of 2.3% in each case. Squamous cell carcinomas (SCCs) occurred in 3.4% of patients in the sorafenib group in the DECISION trial (3). Acute myeloid leukemia and bladder cancer 636

were described in 2.1% of cases (3). Only one patient on the sorafenib arm died due to myocardial infarction attributed to the study drug (3). Rare adverse events reported in patients who received lenvatinib were: acute renal failure (any grade, 4.2%; grade ≥ 3, 1.9%), hepatic failure (grade ≥ 3, 0.4%), gastrointestinal fistula (any grade, 1.5%; grade ≥ 3, 0.8%), and posterior reversible encephalopathy syndrome (any grade, 0.4%; grade ≥ 3, 0) (4). The aim of this manuscript is to communicate rare adverse events that occurred in the clinical setting due to the use of these drugs.

SUBJECTS AND METHODS The authors performed a retrospective case-series study. Out of 760 patients with thyroid cancer, followed up in our Division of Endocrinology, 29 (3.8%) received treatment with MKIs. All of them had progressive disease at the time of the MKI initiation. Sorafenib was prescribed as first-line treatment to 23 patients with DTC and as second-line treatment in one patient with Arch Endocrinol Metab. 2018;62/6


Multikinase inhibitors rare adverse events

RESULTS The baseline characteristics of the 29 patients with thyroid cancer who received treatment with MKIs can be observed in Table 1. During the treatment follow-up (mean 13.7 ± 7 months, median 12 months, range 6-56), 5/29 (17.2%) patients presented rare adverse events. The characteristics and outcomes of these 5 patients are presented in Table 2. A KaplanMeier survival curve shows the time between initiation of MKIs and development of rare adverse events. The estimated median rare adverse event-free survival was not reached for our cohort of patients (Figure 1).

Table 1. Baseline characteristics of 29 patients with thyroid cancer who received treatment with multikinase inhibitors (MKIs) Variable

(n = 29)

Gender Male Female

7 (24.1%) 22 (75.9%)

Age at diagnosis of thyroid cancer (years) Median (range)

53 (20-70)

Histology Differentiated thyroid cancer Medullary thyroid cancer

23 (79%) 6 (21%)

First-line treatment Sorafenib Vandetanib

23 (79%) 6 (21%)

Second-line treatment Sorafenib Vandetanib Lenvatinib Duration of MKI treatment (months) Median (range)

Arch Endocrinol Metab. 2018;62/6

1 (3%) 0 (0%) 2 (7%) 12 (6-56)

Heart failure A 49-year-old woman with a poorly differentiated insular thyroid cancer, metastatic to lungs and bones, was treated with sorafenib 800 mg/day. Nine months after sorafenib initiation, we observed a stable disease. She came to our hospital with severe heart failure. During this event, we excluded other common causes of heart failure. The electrocardiogram was also normal. The initial ejection fraction (EF) before sorafenib prescription was 67%, which decreased to 25% when heart failure developed. One month after sorafenib withdrawal, the EF increased to 55%. The patient did not receive any other MKI treatment. However, she died 5 months later; it was a sudden death. This patient did not receive any other medication prior to the event that could have precipitated the heart failure or arrhythmia leading to sudden death.

Thrombocytopenia A 67-year-old woman developed dysphagia and dyspnea 63 months after initial treatment of papillary thyroid cancer. An 18-fluorodeoxyglucose (FGD) PET/ computerized tomography showed progression of a local mass together with pulmonary metastases. Due to local unresectable disease, sorafenib 800 mg/day was then prescribed. Four months after sorafenib initiation, a grade 3 thrombocytopenia (25,000/mm3) developed and sorafenib was discontinued. The patient presented subconjunctival hemorrhage with spontaneous resolution. No other bleeding manifestations were detected. We performed a bone marrow study to exclude other causes of thrombocytopenia, but no other bone marrow cell linage reductions were observed. Two weeks after sorafenib withdrawal, the platelet count was again in the normal range. Sorafenib was restarted at 400 mg/day, and no evidence of thrombocytopenia was observed after 30 months of treatment with a partial response to sorafenib treatment, according to RECIST 1.1 criteria.

Squamous cell carcinoma (SCC) Two patients developed SCC. The first patient was a 70-year-old man with a diagnosis of advanced and progressive papillary thyroid carcinoma with pulmonary metastases with partial response to sorafenib treatment. Twenty-two months after the MKI initiation, he developed SCC on his back, and 23 months later, three new lesions (two in the forearm and one in the ear) 637

Copyright© AE&M all rights reserved.

advanced MTC. Vandetanib was indicated as first-line treatment in 6 patients with MTC and lenvatinib (which is currently available only under the compassionate use program in our country) as second-line treatment in two patients with progressive disease under sorafenib treatment. All patients undergoing MKI therapy followed an assessment protocol including 12-lead electrocardiogram parameters, vital signs, clinical chemistry, hematology, and urinalysis performed at 1, 2, 4, 8, and 12 weeks and every 3 months thereafter. Adverse events were assessed using the National Cancer Institute’s Common Terminology Criteria for Adverse Events (CTCAE, v5.0).


Multikinase inhibitors rare adverse events

were also diagnosed as SCC. All of them were surgically removed. Currently, 56 months after sorafenib initiation, he continues the treatment at 400 mg/day dosage with partial response to treatment and with strict dermatologic control.

The second case is a 64-year-old patient with advanced MTC. He initially underwent cervical surgery three times (total thyroidectomy with central and left lateral lymphadenectomy, right lateral lymphadenectomy, and mediastinum lymphadenectomy). Three years after the

Table 2. Characteristics and outcomes of 6 patients with rare adverse events (AEs) occurring after multikinase inhibitor (MKI) treatment

1

Age, gender

AE

Histology

MKI

Time after initiation before developing the AE (months)

49, F

Cardiomyopathy (CHF)

Insular

Sorafenib

9

Improvement after suspension

Inhibition of VEGFR and PDGFR-β

Sorafenib

4

Recovery after withdrawal (800 mg/day) and reinitiation with 400 mg/day

Bone marrow affection secondary to RAI

Vandetanib Sorafenib

15 1.5

Surgery

Unknown Inhibition of VEGFR

Outcome

Mechanism that was probably involved

2

67, F

Thrombocytopenia

Classic Papillary

3

64, M

SCC Diverticulitis and intestinal perforation

Medullary

4

70, M

4 SCC (4 surgical resections)

Classic Papillary

Sorafenib

22

Surgery

Activation of MAPK pathway

5

37, F

Oophoritis and intestinal perforation

Medullary

Vandetanib

12

Unilateral salpingooophorectomy

Inhibition of VEGFR

F: female; M: male; CHF: congestive heart failure; SCC: squamous cell carcinoma; VEGFR: vascular endothelial growth factor receptor; PDGFR-β: platelet-derived growth factor receptor β; MAPK: mitogen-activated protein kinase; RAI: radioiodine.

1.0

Rare adverse event-free survival (%)

0.8

0.6

0.4

Copyright© AE&M all rights reserved.

0.2

0.0 0

20

40 Time from initiation of MKI (months)

60

80

Figure 1. Kaplan-Meier curve showing the time between initiation of multikinase inhibitor (MKI) and development of rare adverse events. 638

Arch Endocrinol Metab. 2018;62/6


initial treatment, cervical radiotherapy (4500 cGy) was indicated due to the presence of esophagus infiltration, but 5 years later, progression of mediastinum lymph nodes and hepatic and pulmonary metastases occurred. The largest pulmonary node was 2.2 cm in size and was located in the subpleural region of the right lower lobe. Thus, vandetanib 300 mg/day was prescribed. After 15 months of treatment, two skin tumors with rapid progression were detected, one over his chest and the other over his outer ear. The biopsy showed SCC. After 21 months of vandetanib treatment, he developed progressive disease and sorafenib 800 mg/ day was prescribed as a second-line treatment. Forty days later, sorafenib was interrupted due to evidence of acute diverticulitis. The patient was then hospitalized due to a colonic perforation. Two months later, the patient died from generalized sepsis.

Oophoritis and gastrointestinal perforation A 37-year-old woman was diagnosed with MTC. Sixteen years after the initial diagnosis, she developed distant metastases in her liver and lungs associated with ectopic Cushing's syndrome. Therefore, vandetanib 300 mg/day was prescribed. After 1 month of treatment, she normalized the 24-hour urinary-free cortisol with improvement of signs and symptoms of Cushing's syndrome. She continued with the treatment without complications, but 1 year later, she presented at the emergency department with acute abdominal pain secondary to an ileal perforation. The pathological examination also described the presence of salpingooophoritis. Vandetanib was then stopped, and the Cushing's syndrome recurred immediately. She died 3 months after vandetanib withdrawal due to sepsis.

DISCUSSION A multidisciplinary team is always necessary for the prescription of MKIs in order to monitor and manage the adverse events that will inexorably occur in the majority of patients. In the DECISION trial, the most commonly reported adverse effects in sorafenib treated patients were hand foot syndrome, diarrhea, alopecia, rash, weight loss, and hypertension (3). In the ZETA trial, diarrhea, rash, nausea, and hypertension occurred in more than 30% of patients receiving vandetanib (2). Adverse effects occurring in less than 5% of patients in prospective trials are usually considered rare (2-4). Arch Endocrinol Metab. 2018;62/6

A serious and infrequent adverse event associated with MKI prescription is systolic and diastolic congestive heart failure (5). Patients may present with very dramatic symptoms of heart failure, as occurred with one of our patients (Case 1). This toxicity is not completely understood, but platelet-derived growth factor receptor-β (PDGFR-β) and vascular endothelial growth factor (VEGF) pathway inhibition (6) has been implicated as playing a role in the response to pressure overload-induced stress (7). The patient in Case 1 developed systolic congestive heart failure 7 months after the initiation of sorafenib therapy with improvement of the EF after sorafenib withdrawal. The meta-analysis of Qi and cols. (8) including 10,553 patients with cancer treated with VEGFRMKIs showed an incidence of all-grade and high-grade congestive heart failure of 3.2% and 1.4%, respectively. Our patient eventually presented a sudden death after sorafenib withdrawal, which was probably not related to the MKI treatment. Sudden death was reported in only one case in the meta-analysis performed by Schutz and cols. (9). Interestingly, a fatal case of cardiac failure after 14 months of vandetanib treatment was reported by Scheffel and cols. (10). In this report, myocardial infarction, coronary artery disease, Chagas’s disease, and myocarditis was ruled out, and the postmortem histopathologic findings showed signs suggestive of chronic cardiotoxicity associated with VEGFR inhibitors (10). Thrombocytopenia is another rare adverse effect presented in patients being treated with sorafenib. In hepatocellular carcinoma patients (SHARP study), the frequency of grade 3-4 thrombocytopenia was only 4% for those treated with sorafenib versus 1% for the placebo group (11), and in renal cell carcinoma patients (TARGET Study), this adverse effect occurred in only 1% of those treated with sorafenib versus 0% for the placebo group (12). In the DECISION trial, the frequency of thrombocytopenia was 18.4% for the sorafenib arm versus 9.6% in the placebo group. No grade 3 or 4 events were reported (3). The patient in Case 2 presented with grade 3 thrombocytopenia, which was solved with a dose reduction of the drug. On the other side, secondary malignancies were reported in 4.3% of patients treated with sorafenib in the DECISION study, including 7 patients with skin SCC (3). We reported a patient who developed skin SCC during sorafenib treatment. Sorafenib seems to promote keratinocyte alteration and proliferation 639

Copyright© AE&M all rights reserved.

Multikinase inhibitors rare adverse events


Multikinase inhibitors rare adverse events

through activation of the mitogen-activated protein kinase pathway and BRAF/CRAF heterodimerization, with subsequent activation of CRAF (13). Contrary to sorafenib, vandetanib was not associated with skin SCC. Vandetanib works by blocking RET, VEGFR-2, VEGFR-3, and EGFR (14). Indeed, the inhibition of EGFR and VEGFR has been implicated as a therapeutic target for head and neck SCC (15). However, although we do not believe this is the case, the possibility that the skin SCC in the patient from Case 5 was related to the vandetanib treatment cannot be completely excluded. Finally, we reported the case of a patient who presented an intestinal perforation and salpingo-oophoritis during vandetanib treatment. The risk of gastrointestinal perforation has been associated with VEGFR inhibition, leading to vasoconstriction and thrombosis (16). However, in a systematic review including 300 patients under vandetanib treatment, the use of VEGFR tyrosine kinase inhibitors did not significantly increase the risk of gastrointestinal perforation in comparison with controls (OR 2.99, 95% CI 0.85-10.58, p = 0.089) (16). In conclusion, we present 5 case reports of patients who developed rare (less than 5% in terms of frequency in clinical trials) adverse effects under sorafenib (n = 3) and vandetanib (n = 2) treatment. About 3 to 5 years after their approval, we have learned that these drugs usually lead to adverse events in the majority of patients. Although most of them are manageable, we still need to be aware of potentially serious and rare or unreported adverse effects that can also be lifethreatening. The rapid contact with the treating medical team is of vital relevance to prevent worse outcomes. Disclosure: FabiĂĄn Pitoia is medical advisor, speaker and Steering Committee Bayer. Consultancy for Sanofi and Raffo Laboratories. The rest of the authors have no conflicts of interests to declare.

2. Wells SA, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J Clin Oncol. 2012;30(2):134-41. 3. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, doubleblind, phase 3 trial. Lancet. 2014;384(9940):319-28. 4. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372(7):621-30. 5. Cabanillas ME, Hu MI, Durand JB, Busaidy NL. Challenges associated with tyrosine kinase inhibitor therapy for metastatic thyroid cancer. J Thyroid Res. 2011;2011:985780. 6. Kamba T, McDonald DM. Mechanisms of adverse effects of antiVEGF therapy for cancer. Br J Cancer. 2007;96(12):1788-95. 7. Chintalgattu V, Ai D, Langley RR, Zhang J, Bankson JA, Shih TJ, et al. Cardiomyocyte PDGFR-beta signaling is an essential component of the mouse cardiac response to load-induced stress. J Clin Invest. 2010;120(2):472-84. 8. Qi WX, Shen Z, Tang LN, Yao Y. Congestive heart failure risk in cancer patients treated with vascular endothelial growth factor tyrosine kinase inhibitors: a systematic review and meta-analysis of 36 clinical trials. Br J Clin Pharmacol. 2014;78(4):748-62. 9. Schutz FAB, Je Y, Richards CJ, Choueiri TK. Meta-analysis of randomized controlled trials for the incidence and risk of treatmentrelated mortality in patients with cancer treated with vascular endothelial growth factor tyrosine kinase inhibitors. J Clin Oncol. 2012;30(8):871-7. 10. Scheffel RS, Dora JM, Siqueira DR, Burttet LM, Cerski MR, Maia AL. Toxic cardiomyopathy leading to fatal acute cardiac failure related to vandetanib: a case report with histopathological analysis. Eur J Endocrinol. 2013;168(6):K51-4. 11. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378-90. 12. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356(2):125-34. 13. Fathi AT, Lin WM, Durazzo T, Piris A, Sadrzadeh H, Bernardo L, et al. Extensive squamous cell carcinoma of the skin related to use of sorafenib for treatment of FLT3-mutant acute myeloid leukemia. J Clin Oncol. 2016;34(20):70-2. 14. Fallahi P, Di Bari F, Ferrari SM, Spisni R, Materazzi G, Miccoli P, et al. Selective use of vandetanib in the treatment of thyroid cancer. Drug Des Devel Ther. 2015;9:3459-70.

1. Haugen BR, Alexander EK, Bible KC, Doherty G, Mandel SJ, Nikiforov YE, et al. American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer. Thyroid. 2016;26(1):1-133.

16. Qi WX, Sun YJ, Tang LN, Shen Z, Yao Y. Risk of gastrointestinal perforation in cancer patients treated with vascular endothelial growth factor receptor tyrosine kinase inhibitors: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2014;89(3):394-403.

CopyrightŠ AE&M all rights reserved.

REFERENCES

15. Klein JD, Christopoulos A, Ahn SM, Gooding WE, Grandis JR, Kim S. Antitumor effect of vandetanib through EGFR inhibition in head and neck squamous cell carcinoma. Head Neck. 2012;34(9):1269-76.

640

Arch Endocrinol Metab. 2018;62/6


brief report

Selective screening for thyroid dysfunction in pregnant women: How often do low-risk women cease to be treated following the new guidelines of the American Thyroid Association? Pedro Weslley Rosario1

ABSTRACT Objective: Universal screening for thyroid dysfunction in pregnant women is not recommended by the American Thyroid Association (ATA) or the American Association of Clinical Endocrinologists (AACE). This study evaluated the frequency of pregnant women that would have an indication for levothyroxine (L-T4) according to the new ATA/AACE guidelines among low-risk women without an indication for screening with TSH. Subjects and methods: The sample consisted of 412 pregnant women ranging in age from 18 to 30 years. These women were considered to be at low risk for thyroid dysfunction according to ATA/AACE and would not be candidates for screening with TSH. Anti-thyroid peroxidase antibodies (TPOAb) and TSH were measured. Women who had TSH > 2.5 mIU/L or TPOAb in the first trimester were submitted to subsequent evaluations in the second and third trimester. Results: In the first trimester, none of the pregnant women would have L-T4 therapy “recommended” and treatment would be “considered” in only two. In the second trimester, pregnant women with positive TPOAb or TSH > 2.5 mIU/L in the first trimester (n = 30) were reevaluated. L-T4 treatment would be “recommended” in only one woman and would be “considered” in two others. The 28 women that were not treated in the second trimester were reevaluated in the third trimester, but none of them would have L-T4 “recommended”. Conclusion: The findings of the study suggest that selective screening, recommended by ATA/AACE does not result in a significant loss of pregnant women with an indication for L-T4 treatment. Arch Endocrinol Metab. 2018;62(6):641-3

Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brasil 1

Correspondence to: Pedro Weslley Rosario Instituto de Ensino e Pesquisa, Santa Casa de Belo Horizonte Rua Domingos Vieira, 590 30150-240 – Belo Horizonte, MG, Brasil pedrowsrosario@gmail.com Received on June/30/2018 Accepted on Nov/6/2018 DOI: 10.20945/2359-3997000000089

INTRODUCTION

T

he most recent guidelines of the American Thyroid Association (ATA), reviewed and endorsed by the American Association of Clinical Endocrinologists (AACE), recommend that: (i) when indicated, screening for thyroid dysfunction in pregnancy should be performed during the first prenatal visit, which generally occurs in the first trimester; (ii) this screening starts with the measurement of serum TSH, and (iii) TSH between 0.1 and 2.5 mIU/L closes the investigation (1). Universal screening for thyroid dysfunction in pregnant women is not recommended by ATA/ AACE (1). The recommendation is that only high-risk pregnant women be investigated by TSH measurement (1). The definition of this group was also reinforced in the recent guidelines (1). Although several conditions are considered risk conditions, many argue that the selective screening strategy (1) fails to diagnose thyroid Arch Endocrinol Metab. 2018;62/6

dysfunction in a significant portion of low-risk pregnant women, thus recommending universal screening (2,3). Controversy regarding screening for thyroid dysfunction in pregnant women also exists in Brazil. In the consensus on “the clinical use of thyroid function tests”, in addition to individuals with risk factors, pregnancy is an indication for TSH measurement (4). On the other hand, the consensus on subclinical hypothyroidism concluded that “there is insufficient evidence to recommend or not recommend universal screening for hypothyroidism with TSH in pregnant women in the first trimester of gestation” (5). This divergence is also observed in clinical practice. A Latin American study showed that 38.4% of responders use a universal screening strategy and 43% prefer a casefinding approach in high-risk groups (6). Finally, the recommendations for levothyroxine (L-T4) treatment were revised in the recent ATA/AACE guidelines (1). 641

Copyright© AE&M all rights reserved.

Keywords Screening; thyroid dysfunction; pregnancy


Thyroid dysfunction in pregnant women

To our knowledge, no Brazilian study has evaluated the frequency of pregnant women that would have an indication for L-T4 treatment according to the new ATA/AACE guidelines (1) among women with low risk and therefore without an indication for screening with TSH (1). This was the objective of the present study.

SUBJETCS AND METHODS The study was approved by the local Research Ethics Committee (7,8). The population studied was from the metropolitan region of Belo Horizonte (Minas Gerais, Brazil), an area where iodine intake is adequate. Nine hundred and ninety-two pregnant women with ≤ 12 weeks gestation who underwent prenatal tests at a clinical analysis laboratory and who had become pregnant spontaneously were initially interviewed and examined (7,8). For this study, women who met the clinical criteria shown in Table 1 (n = 480) were excluded. The sample consisted of 412 women ranging in age from 18 to 30 years, with a median gestation of 9 weeks, including 212 primigravidae. These women were at low risk for thyroid dysfunction according to ATA/AACE and would not be candidates for screening with TSH (1). Anti-thyroid peroxidase antibodies (TPOAb) and TSH were measured. Only women who had TSH > 2.5 mIU/L or positive TPOAb in the first trimester were submitted to subsequent evaluations in the second and third trimester. Regarding L-T4 therapy, two categories were defined according to the ATA/AACE guidelines (1): (i) therapy “recommended”, if TSH > 10 mIU/L or TSH entre 4 and 10 mIU/L with positive TPOAb, Table 1. Exclusion criteria Known thyroid disease, current or previous treatment with antithyroid drugs or L-T4 History of 131I therapy, thyroidectomy or head and neck external radiotherapy Age > 30 years

and (ii) therapy “considered”, if TSH between 4 and 10 mIU/L without TPOAb or TSH between 2.5 and 4 mIU/L with positive TPOAb. Serum samples were obtained from the women in the morning (at about 8 a.m.) after an 8- to 10-h fast. TSH was measured with a chemiluminescent assay (Immulite 2000, Diagnostic Products Corporation, Los Angeles, CA), with reference values of 0.4-4 mIU/L. TPOAb were also measured with a chemiluminescent assay (Immulite 2000), with reference values of up to 35 kIU/L.

RESULTS In the first assessment (first trimester), none of the pregnant women would have L-T4 therapy “recommended” and treatment would be “considered” in only two. These women had positive TPOAb and TSH between 2.82 and 3.12 mIU/L. None of the women was treated with L-T4 in this first assessment. Of note, none of the women would be a candidate for antithyroid drug treatment [TSH < 0.1 mIU/L with elevated free T4 and positive anti-TSH receptor antibodies (TRAb) (1)]. Pregnant women with positive TPOAb (n = 10) or TSH > 2.5 mIU/L (n = 18) or both (n = 2) in the first assessment were reevaluated in the 22nd week of gestation (second trimester). L-T4 treatment would be “recommended” in only one woman (she had TSH 4.8 mIU/L and positive TPOAb) who was actually treated. In other two patients, L-T4 therapy would be “considered”, one with TSH 3.5 mIU/L and positive TPOAb and the other with TSH 4.5 mIU/L without TPOAb. L-T4 was initiated only in the first patient. The 28 women who were not treated in the second trimester were reevaluated in the 34th week of gestation (third trimester), but none of them would have L-T4 treatment “recommended” (1). The woman for whom therapy would be “considered” in the second trimester (1) continued with this classification in the third trimester.

Type 1 diabetes or other autoimmune diseases History of pregnancy loss, preterm delivery, or infertility Copyright© AE&M all rights reserved.

Multiple prior pregnancies (≥ 2) Family history of autoimmune thyroid disease or thyroid dysfunction Morbid obesity (BMI ≥ 40 kg/m2) Use of amiodarone, interferon, or lithium; or recent (in the past 8 weeks) exposure to iodinated contrast agents Goiter, palpable thyroid anomaly or ophthalmopathy

642

DISCUSSION Considering that pregnant women with TSH < 2.5 mIU/L without TPOAb would remain without an indication for L-T4 therapy throughout pregnancy (1) and applying the recommendations of the new ATA/ AACE guidelines (1), among the 412 women at low Arch Endocrinol Metab. 2018;62/6


Thyroid dysfunction in pregnant women

risk for thyroid dysfunction (1), only one would have treatment “recommended” and treatment would be “considered” in two (1). Population differences may exist, and it is possible that our results cannot be reproduced in all populations. Furthermore, pregnant women with TSH < 2.5 mIU/L without TPOAb were not reevaluated in the second or third trimester, but these women do not require additional investigation according to ATA/AACE (1). In addition, since these women did not have TPOAb, L-T4 treatment would only be “considered” if TSH exceeds 4 mIU/L and only above 10 mIU/L would treatment be “recommended” (1), which we believe is highly unlikely to occur. Despite these limitations, the study included a reasonable number of pregnant women who were rigorously selected and considered to be at low risk for thyroid dysfunction following the definition of ATA/AACE (1). In addition, the treatment indications were evaluated according to the most recent guidelines of ATA/AACE (1). The findings of the study suggest that selective screening, recommended by ATA/AACE (1), does not result in a significant loss of pregnant women with an indication for L-T4 treatment. Since in the present series low-risk pregnant women accounted for less than half of the initial sample, the high-risk definition proposed by ATA/AACE (1) seems to be poorly selective but has a high negative predictive value.

Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Alexander EK, Pearce EN, Brent GA, Brown RS, Chen H, Dosiou C, et al. 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017;27:315-89. 2. Pop VJ, Broeren MA, Wiersinga WM, Stagnaro-Green A. Thyroid disease symptoms during early pregnancy do not identify women with thyroid hypofunction that should be treated. Clin Endocrinol (Oxf). 2017;87:838-43. 3. Stagnaro-Green A. Clinical guidelines: Thyroid and pregnancy time for universal screening? Nat Rev Endocrinol. 2017;13:192-4. 4. Carvalho GA, Perez CL, Ward LS. The clinical use of thyroid function tests. Arq Bras Endocrinol Metabol. 2013;57:193-204. 5. Sgarbi JA,Teixeira PF, Maciel LM, Mazeto GM, Vaisman M, Montenegro Junior RM, et al.; Brazilian Society of Endocrinology and Metabolism. The Brazilian consensus for the clinical approach and treatment of subclinical hypothyroidism in adults: recommendations of the thyroid Department of the Brazilian Society of Endocrinology and Metabolism. Arq Bras Endocrinol Metabol. 2013;57:166-83. 6. Medeiros MF, Cerqueira TL, Silva Junior JC, Amaral MT, Vaidya B, Poppe KG, et al.; Latin AmericanThyroid Society. An international survey of screening and management of hypothyroidism during pregnancy in Latin America. Arq Bras Endocrinol Metabol. 2014;58:906-11. 7. Rosario PW, Carvalho M, Calsolari MR. TSH reference values in the first trimester of gestation and correlation between maternal TSH and obstetric and neonatal outcomes: a prospective Brazilian study. Arch Endocrinol Metab. 2016;60:314-8. 8. Rosario PW, Oliveira LFF, Calsolari MR. Maternal hypothyroxinemia in the first trimester of gestation and association with obstetric and neonatal outcomes and iron deficiency: a prospective Brazilian study. Arch Endocrinol Metab. 2018;62:332-6.

Copyright© AE&M all rights reserved.

Funding: this work was supported by the National Council for Scientific and Technological Development (CNPq).

Compliance with ethical standards: the study was approved by the Research Ethics Committee of our institution.

Arch Endocrinol Metab. 2018;62/6

643


case report

Divisão de Cirurgia Pediátrica, Departamento de Cirurgia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCMUnicamp), Campinas, SP, Brasil 2 Grupo Interdisciplinar de Estudos da Determinação e Diferenciação do Sexo (GIEDDS), Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCM-Unicamp), Campinas, SP, Brasil 3 Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCM-Unicamp), Campinas, SP, Brasil 4 Departamento de Genética Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCM-Unicamp), Campinas, SP, Brasil 5 Departamento de Pediatria, Unidade de Endocrinologia Pediátrica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCMUnicamp), Campinas, SP, Brasil * SCC and JGRA contributed equally to this work 1

Early development of a gonadal tumor in a patient with mixed gonadal dysgenesis Sarah Crestian Cunha1*, Juliana Gabriel Ribeiro de Andrade2*, Camila Matsunaga de Angelis3, Athanase Billis3, Joaquim Murray Bustorff-Silva1, Andréa Trevas Maciel-Guerra2,4, Márcio Lopes Miranda1,2, Gil Guerra-Júnior2,5

SUMMARY A gonadal tumor was diagnosed in the first months of life in a patient with genital ambiguity, a 45,X/46,XY karyotype, and mixed gonadal dysgenesis. Gonadal biopsies at the age of 3 months revealed dysgenetic testes and a gonadoblastoma on the right testis. Even though gonadal tumors are rare in childhood, this case indicates that prophylactic removal of dysgenetic gonads should be performed as early as possible, especially when the female sex is assigned to a patient with a Y-chromosome sequence. Arch Endocrinol Metab. 2018;62(6):644-7

Correspondence to: Gil Guerra-Júnior Rua Tessália Vieira de Camargo, 126 13083-887 – Campinas, SP, Brasil gilguer@fcm.unicamp.br Received on Apr/30/2017 Accepted on Dec/2/2017 DOI: 10.20945/2359-3997000000091

INTRODUCTION

Copyright© AE&M all rights reserved.

D

isorders of sex development (DSD) are a group of congenital conditions in which the chromosomal, gonadal, or anatomical sex is atypical. These conditions include disorders of gonadal development, comprising various types of gonadal dysgenesis (GD) (1). A GD may be either complete (CGD), with bilateral streak and female internal and external genitalia in XX or XY individuals; partial (PGD), characterized by genital ambiguity and two dysgenetic testes or a dysgenetic testis and a streak gonad in 46,XY subjects; or mixed (MGD), when a 45,X/46,XY karyotype or its variants are found in a patient whose gonadal and genital phenotype is similar to that of PGD (2). In MGD, the presence of a 45,X cell line is associated with features of Turner syndrome, including short stature, cardiac and renal malformations, and thyroid disease, among others; thus, MGD and PGD have different prognoses (3,4). 644

The presence of Y-chromosome sequences in patients with GD increases their risk of developing gonadal tumors (5,6). In patients with MGD, this risk is estimated to be 3-4% and 10-20% at the ages of 10 and 15 years, respectively. The overall risk is 20-25% and may rise up to 46% at the age of 40 years (7). In patients with MGD reared as girls, bilateral gonadectomy should be performed in childhood or upon diagnosis because of the risk of malignancy and insufficient gonadal function. For those reared as boys, removal of streak gonads and strikingly dysgenetic testes should also be done early; testes with lesser degrees of dysgenesis may be preserved in the scrotum and closely followed up for early detection of tumor development (8-11). The aim of this report is to describe a patient with MGD who developed a gonadoblastoma within the first months of life. This case can add knowledge to support the decision about early prophylactic gonadectomy in this group of patients. Arch Endocrinol Metab. 2018;62/6


Gonadal tumor in a patient with mixed GD

CASE REPORT

Arch Endocrinol Metab. 2018;62/6

Figure 1. Biopsy of the right gonad showing germ cells intermingled with seminiferous tubules with decreased diameter.

Figure 2. Large clear germ cells with prominent nuclei interspersed by other small cells with hyperchromatic nuclei and scant cytoplasm (immature Sertoli cells).

Copyright© AE&M all rights reserved.

A 43-day-old infant without sex assignment was referred to us due to genital ambiguity. This infant was born at term by cesarean section and without neonatal complications, with a birth weight of 2,580 g and a length of 45 cm. The infant – the only child of healthy, unrelated parents – also had a healthy, 17-month-old maternal half-brother. The physical examination revealed a healthy infant with normal growth and without dysmorphic features, except for those of the external genitalia. A 3.5-cm phallus with chordee and penoscrotal hypospadias was present, and both gonads were palpable in the inguinal region. The features of the external genitalia were compatible with Prader grade III/IV (12), and the external masculinization score (EMS) (13) was 6.0. The infant’s karyotype was 45,X[7]/46,XY[43] and, on pelvic ultrasound, uterus and vagina were visualized. Basal levels of gonadotropins (FSH: 4.87 IU/L; LH: 5.45 IU/L), total testosterone (2.51 ng/mL), androstenedione (8.76 mg/dL), estradiol (22 pg/mL), and 17-hydroxyprogesterone (216.6 ng/dL) were normal for the reference values at minipuberty (when the hormonal values are similar to those of normal puberty). Abdominal ultrasound showed no urinary tract malformations, and echocardiography was also normal. Bilateral inguinotomy and biopsies of both gonads were performed at the age of 3 months to evaluate the degree of GD or the presence of ovarian tissue. At the same time, laparoscopy – which is the gold standard for the evaluation of internal genitalia (14) – confirmed the presence of uterus and fallopian tubes. The histology revealed bilateral dysgenetic testes with signs of neoplastic transformation on the right gonad (Figure 1). Based on the diagnosis of MGD, the anatomy of the internal and external genitalia, and the wish of the parents, the female gender was assigned to the infant, and bilateral gonadectomy was performed. The histological evaluation showed signs of bilateral testicular dysgenesis (areas of undifferentiated gonadal tissue, immature tubules surrounded by fibrosis together with well-differentiated testicular tissue, intracapsular tubules). The right gonad had two neoplastic lesions: a premalignant one (germ cell cluster, clonal proliferation), within a region of undifferentiated gonadal tissue, and a gonadoblastoma (Figures 2 and 3).

Figure 3. Germ cells positive for OCT3/4. 645


Gonadal tumor in a patient with mixed GD

When the infant was 12 months old, a feminizing genitoplasty was successfully performed. She is currently 4 years old, well-adapted to the female social gender, and has a normal growth and neurological development.

DISCUSSION The finding of a 45,X/46,XY karyotype in patients with genital ambiguity and a female internal genitalia opens up the possibility for two differential diagnoses: MGD or ovotesticular DSD (1). The absence of ovarian tissue and the presence of bilateral dysgenetic testes in the patient presented here confirmed the diagnosis of MGD (1). Even though in the past patients with bilateral testicular dysgenesis have been histologically classified as having PGD (2,5), the classification is now based on the infant’s karyotype, and patients with a 45,X cell line are now diagnosed as having MGD (1,3). This classification is undoubtedly more useful, because the presence of a 45,X lineage prompts the evaluation of features of Turner syndrome: short stature, associated congenital anomalies (heart, kidney, and urinary system), as well as other associated diseases (autoimmune thyroid disease and hearing loss, among others) (3). The use of histological features for the classification may be misleading, due to the frequent limitation in evaluating the entire gonad. Therefore, the diagnosis of MGD in this case is in line with the DSD Consensus (1). The most relevant aspect of this report is the presence of a gonadoblastoma in the right gonad, which was initially suspected at the age of 3 months and confirmed after the removal of the gonads. As of now, only a few cases of gonadal tumors have been described in very young patients with testicular dysgenesis. These include a carcinoma in situ in a 2-year-old boy with ambiguous genitalia, a 45,X/46,XY karyotype, and a dysgenetic

right inguinal testis (4); bilateral gonadoblastoma in a 3-year-old girl with female genitalia, 46,XY karyotype, and bilateral dysgenetic gonads (6); and a left abdominal mass compatible with seminoma in a 15-day-old girl with ambiguous genitalia, 46,XY karyotype, and a right testis (15). In addition, bilateral gonadoblastoma has also been reported in a 7-month-old 46,XY girl with mutations in the WT1 gene, dysgenetic right testis, and chronic renal failure in the first year of life; the original histology of the left gonad in the patient was undetermined (16). Despite the usual benign behavior of gonadoblastomas, malignant transformation to seminomatous and nonseminomatous tumors can occur over the years. Other types of germ cell tumors associated with gonadoblastomas may be metastatic: in half of the cases, the tumor cells invade the stroma and turn into a dysgerminoma or seminoma (8). About 26% of all gonadoblastomas arise from dysgenetic gonads, and 20% of them emerge from cryptorchid dysgenetic testes, but in 54% of cases, the original structure of the gonad is hidden by the tumor (9). Recently, Pleskacova and cols. (10) have defined risk groups for the development of gonadal tumors in GD according to the gonadal location, histopathology, and immunohistochemistry, as shown in Table 1. Concerning the present case, although the gonad was within the inguinal canal and the child was less than 1 year old, there were other features compatible with high-risk criteria for malignancy (ambiguous genitalia, dysgenetic testis, and positive and focal OCT3/4). In conclusion, this rare case with an early neoplastic transformation confirms the need to carry out prophylactic gonadectomy in MGD patients as early as possible, particularly when the female gender is assigned to the infant. Disclosure: no potential conflict of interest relevant to this article was reported.

Table 1. Groups with dysgenetic gonads at risk for the development of gonadal tumors (10)

CopyrightŠ AE&M all rights reserved.

Low

Medium

High

Degree of virilization

Male genitalia

Slight undervirilization

Ambiguous genitalia

Gonadal location

Scrotal

Inguinal

Abdominal

Histopathology

Dysgenetic gonad without germ cell or ovary or dysgenetic testis with negative OCT3/4

Dysgenetic testis positive OCT3/4

Undifferentiated gonadal tissue or dysgenetic testicle positive OCT3/4

Negative OCT3/4

Positive OCT3/4 dispersed in the gonad, negative or weakly positive TSPY, negative stem cell factor, age less than 1 year

Positive OCT3/4 located in the basal lamina, focal location, strongly positive TSPY, positive stem cell factor, age more than 1 year

Immunohistochemistry

646

Arch Endocrinol Metab. 2018;62/6


Gonadal tumor in a patient with mixed GD

REFERENCES 1. Lee PA, Houk CP, Ahmed SF, Hughes IA; International Consensus Conference on Intersex organized by the Lawson Wilkins Pediatric Endocrine Society and the European Society for Paediatric Endocrinology. Consensus statement on management of intersex disorders. International Consensus Conference on Intersex. Pediatrics. 2006;118(2):e488-500. 2. McCann-Crosby B, Mansouri R, Dietrich JE, McCullough LB, Sutton VR, Austin EG, et al. State of the art review in gonad dysgenesis: challenges in diagnosis and management. Int J Pediatr Endocrinol. 2014;2014(4):1-17. 3. Andrade JG, Guerra-Júnior G, Maciel-Guerra AT. 46,XY and 45,X/46,XY testicular dysgenesis: similar gonadal and genital phenotype, different prognosis. Arq Bras Endocrinol Metabol. 2010;54(3):331-4. 4. Johansen ML, Hagen CP, Rajpert-De-Meyts E. 45,X/46,XY mosaicism: phenotypic characteristics, growth, and reproductive function – a retrospective longitudinal study. J Clin Endocrinol Metab. 2012;97:e1540-9. 5. Cools M, Drop SLS, Wolffenbuttel KP, Oosterhuis JW, Looijenga LHJ. Germ cell tumors in the intersex gonad: old paths, new directions, moving frontiers. Endocr Rev. 2006;27(5):468-84. 6. Wunsch L, Holterhus PM, Wessel L. Patients with disorders of sex development (DSD) at risk of gonadal development based on laparoscopic biopsy and molecular diagnosis. BJU Int. 2012;110:e958-65.

9. Cools M, Looijenga LH, Wolffenbuttel KP, Drop SL. Disorders of sex development: update on the genetic background, terminology and risk for development of germ cell tumors. World J Pediatr. 2009;5(2):93-102. 10. Pleskacova J, Hersmus R, Oosterhuis JW, Setyawati BA, Faradz SM, Cools M, et al. Tumor risk in disorders of sex development. Sex Dev. 2010;4(4-5):259-69. 11. Lee PA, Nordenström A, Houk CP, Ahmed SF, Auchus R, Baratz A, et al. Global Disorders of Sex Development Update since 2006: Perceptions, Approach and Care. Horm Res Paediatr. 2016;85(3):158-80. 12. Prader A. Genital findings in the female pseudo-hermaphroditism of the congenital adrenogenital syndrome; morphology, frequency, development and heredity of the different genital forms. Helv Paediatr Acta. 1954;9(3):231-48. 13. Ahmed SF, Khwaja O, Hughes IA. The role of a clinical score in the assessment of ambiguous genitalia. BJU Int. 2000;85(1):120-4. 14. Guerra-Junior G, Andrade KC, Barcelos IHK, Maciel-Guerra AT. Imaging techniques in the diagnostic journey of Disorders of Sex Development. Sex Dev. 2018;12(1-3):95-99. 15. Robboy SJ, Miller T, Donahoe PK, Jare C, Welch WR, Haseltine FP, et al. Dysgenesis of testicular and streak gonads in the syndrome of mixed gonadal dysgenesis: perspective derived from a clinicopathologic analysis of twenty-one case. Hum Pathol. 1982;13(8):700-16. 16. Patel PR, Pappas J, Arva NC, Franklin B, Brar PC. Early presentation of bilateral gonadoblastomas in a Denys-Drash syndrome patient: a cautionary tale for prophylactic gonadectomy. J Pediatr Endocrinol Metab. 2013;26(9-10):971-4.

Copyright© AE&M all rights reserved.

7. Abaci A, Çatli G, Berberoglu M. Gonadal malignancy risk and prophylactic gonadectomy in Disorders of sexual development. J Pediatric Endocrinol Metab. 2015;28(9-10):1019-27.

8. Oosterhuis JW, Looijenga LH. Testicular germ cell tumors in a broader perspective. Nat Rev Cancer. 2005;5(3):210-22.

Arch Endocrinol Metab. 2018;62/6

647


case report

Suprasellar chordoid glioma: a report of two cases Karina Danilowicz1, Santiago Gonzalez Abbati1, Soledad Sosa1, Florencia Lustig Witis1, Gustavo Sevlever2

SUMMARY Hospital de Clínicas, Universidad de Buenos Aires, Endocrinology, Buenos Aires, Argentina 2 Instituto de Investigaciones Neurológicas “Dr. Raúl Carrea”, FLENI, Pathology, Buenos Aires, Argentina 1

Correspondence to:

Karina Danilowicz kdanilowicz@hotmail.com Received on Feb/26/2016 Accepted on Dec/14/2016 DOI: 10.20945/2359-3997000000092

Chordoid glioma (CG) is considered a slow growing glial neoplasm. We report two new cases with endocrinological presentation, management and outcome. Case reports: 1) An 18 year-old female patient was admitted due to headaches, nausea and vomiting and visual abnormalities. She was in amenorrhea. A brain magnetic resonance imaging (MRI) demonstrated a 35 mm-diameter sellar and suprasellar mass. An emergency ventricular peritoneal valve was placed due to obstructive hydrocephalus. Transcraneal surgery was performed. The patient developed central hypothyroidism, adrenal insufficiency and transient diabetes insipidus; she never recovered spontaneous menstrual cycles. Histopathologic study showed cells in cords, inside a mucinous stroma, positive for glial fibrillary acidic protein (GFAP). Due to residual tumor gamma knife radiosurgery was performed. Three years after surgery, the patient is lucid, with hypopituitarism under replacement. 2) A 46 yearold woman complained about a three year-history of amenorrhea, galactorrhea and headache. An MRI showed a solid-cystic sellar mass 40 mm-diameter that extended to the suprasellar cistern. She had hypogonatropic hypogonadism and mild hyperprolactinemia. The tumor mass was removed via nasal endoscopic approach. Histopathological study reported cellular proliferation of glial lineage positive for GFAP.The patient evolved with central hypothyroidism and diabetes insipidus. She was re-operated for fistula and again under the diagnosis of extradural abscess. She evolved with cardiorespiratory descompensation and death, suspected to be due to a thromboembolism. In conclusion, the first case confirms that best treatment for CG is surgery considering radiotherapy as an adjuvant therapy. The other case, on the contrary, illustrates the potentially fatal evolution due to surgical complications. Arch Endocrinol Metab. 2018;62(6):648-54

INTRODUCTION

C

Copyright© AE&M all rights reserved.

hordoid glioma (CG) is a primary tumor of the central nervous system (1). It was described by Brat et al. in 1998 (2) in a series of 8 cases. In 2007 CG has been included in the World Health Organization (WHO) classification (3) as a neuroepithelial tumor. The incidence is not clear: to our knowledge, there are 99 described cases at the moment. The name CG is due to the histological similarity to chordoma and the immunostaining with glial fibrillary acidic protein (1). It is a rare neoplasm with a chordoid appearance (4), attached to the hypothalamic or suprasellar area. We report two new cases with endocrinological presentation, management and outcome, with a review of the literature.

CASE REPORTS Case 1 An 18 year-old female patient was admitted due to a two-month history of intense headaches, nausea 648

and vomiting. She referred visual abnormalities and progressive sleepiness. She was in amenorrhea for a period of 8 months. She did not refer polyuria. A brain magnetic resonance imaging (MRI) evidenced a sellar and suprasellar expansive mass of 35 mm diameter (Figure 1), with intense homogeneous enhancement on T1 weighted imaging after gadolinium administration and hyperintense on T2 weighted imaging. The mass was hyperdense on computed tomography. Once arrived in our hospital her consciousness worsened and she presented seizures. A ventriculo­ peritoneal shunt plus endoscopic septostomy with biopsy of the third ventricle mass was done at the same procedure, due to obstructive hydrocephalus. The biopsy initially suggested the diagnosis of chordoid glioma versus pilomixoid astrocytoma. Transcranial surgery was performed by a right pterional approach and trans-lamina terminalis partial resection of the tumor was done. Seven days post-surgery she presented left lung atelectasis requiring respiratory assistance. A pulmonary embolism was ruled out. Postoperatively Arch Endocrinol Metab. 2018;62/6


Suprasellar chordoid glioma

A

C

Three months post-surgery she developed poliphagia and memory disturbances. Eighteen months post-surgery, due to residual tumor of 10 per 19 mm (Figure 3), gamma knife radiosurgery was performed. Three years after surgery, the patient is lucid, with a normal appetite, under replacement therapy with levothyroxine 125 Âľg/day, hydrocortisone 15 mg/day and etinilestradiol with levonorgestrel.

B

D

Figure 1. Patient 1. Magnetic resonance imaging findings: sagittal, coronal and axial images. A suprasellar ovoid mass with homogeneous enhancement after gadolinium administration is shown. (A) The sagittal T2 weighted image shows a suprasellar hyperintense ovoid mass with third ventricle extension. Note the normal pituitary gland in a normal sella (arrow). (B) The coronal T1 weighted image shows avid and homogeneous enhancement after gadolinium administration and ventricular asymmetry despite the septostomy and shunt. (C and D) The axial T1 weighted images with gadolinium show the involvement of the tumor in the interpeduncular cistern and the third ventricle. No hydrocephalus was detected due to the ventriculo-peritoneal shunt (asterisks). Arch Endocrinol Metab. 2018;62/6

649

CopyrightŠ AE&M all rights reserved.

the patient developed central hypothyroidism, adrenal insufficiency and transitory diabetes insipidus (DI), and she never recovered spontaneous menstrual cycles. Histopathology showed cells with oval nuclei, in cords, inside a mucinous stroma, positive for glial fibrillary acidic protein (GFAP), and negative for neurofilaments and epithelial membrane antigen (EMA). The diagnosis of chordoid glioma was settled (Figure 2).


Suprasellar chordoid glioma

A

B

CopyrightŠ AE&M all rights reserved.

Figure 2. Immunohistochemical findings. (A) Hematoxylin and eosin stain (400x): small cells with circular and oval nucleus, with eosinophilic cytoplasm in a mucinous stroma. (B) Glial fibrillary acidic protein stain (100x): intense and diffuse reactivity for GFAP, in clusters and cords.

A

B

C

D

Figure 3. Patient 1. Postoperative and pre-radiotherapy magnetic resonance imaging findings. (A) The sagittal T2 weighted image showed remaining tumor at the third ventricle but without no significant mass effect. (B) The coronal T1 gadolinium weighted image showed the tumor inside the third ventricle but with ventricular symmetry. (C) The axial T1 gadolinium weighted image showed remnant tumor in the interpeduncular cistern. (D) The FLAIR sequence showed spontaneous brightness of the chordoid glioma. 650

Arch Endocrinol Metab. 2018;62/6


Suprasellar chordoid glioma

A 46 year-old woman complained about a three yearhistory of amenorrhea and galactorrhea. Headache began one year before consultation, but worsened on the last month. An MRI showed a solid-cystic sellar mass of 22 x 25 x 40 mm, that extended to the suprasellar cistern, interpeduncular and prepontine region. It was homogeneous with gadolinium enhancement on T1 weighted images. The FLAIR and the T2 weighted sequences showed hyperintensity of the lesion. The normal pituitary gland was in situ below the tumor. Mild supratentorial ventricular dilatation was detected (Figure 4). Visual field examination revealed peripheral temporal scotomies in both eyes. She had laboratory abnormalities that evidenced hypogonadotropic hypogonadism and hyperprolactinemia without alterations of thyroid and adrenal axis. With a presumptive diagnosis of papillary craniopharyngioma, the patient was referred for surgical treatment. The tumor mass was in a retrochiasmatic and third ventricle topography, and was removed by extended endonasal endoscopic approach: an extended transplanum transtubercullum sellae approach was done and a total tumor resection could be achieved. Histopathological study reported cellular proliferation of glial lineage with slight nuclear pleomorphism and moderate diffuse eosinophilic cytoplasm. There was sparse myxoid material and mild lymphoplasmocytic infiltrate. Immunostaining was positive for GFAP, with focal expression of EMA in plasma cells. Proliferation A

B

index Ki67 was below 1%. Chordoid glioma was diagnosed (Figure 5). The patient evolved postoperatively with temporal and spatial disorientation, central hypothyroidism and diabetes insipidus. Treatment with levothyroxine and desmopressin was initiated. She developed cerebrospinal fluid fistula and pioventriculitis. Klebsiella pneumoniae was isolated from cerebrospinal fluid. She was re-operated for fistula closure and a temporary external ventricular shunt was placed. Systemic and intrathecal antibiotic treatment was completed with good response. Twenty-eight days after the second intervention, the patient presented fever again and an isointense mass in T1 sequence was observed on MRI, near the place

Figure 5. Immunohistochemical findings. Positive immunostaining for glial fibrillary acidic protein (400 X).

cytoplasmic

C

Figure 4. Patient 2. Magnetic resonance imaging findings: sagittal and coronal images. A suprasellar mass with heterogeneous enhancement after gadolinium administration is shown, with hypothalamic involvement and hydrocephalus. (A) The sagittal T1 weighted image shows a suprasellar tumor with gadolinium enhancement compromising the interpeduncular cistern and the third ventricle. The normal pituitary gland is in a normal position (arrow). (B) The axial T2 weighted image shows spontaneous hyperintensity. (C) The coronal FLAIR shows tumor brightness and associated obstructive hydrocephalus. Arch Endocrinol Metab. 2018;62/6

651

CopyrightŠ AE&M all rights reserved.

Case 2


Suprasellar chordoid glioma

where the fistula was repaired. Under the presumptive diagnosis of extradural abscess, she was submitted for rhinosinusal exploration without microbiological isolation. Sixteen days later, she evolved with cardiorespiratory descompensation, suspected to be due to a thromboembolism, and died.

CopyrightŠ AE&M all rights reserved.

DISCUSSION The chordoid glioma is a rare low-grade neoplasm mostly located in the third ventricle. Obstructive hydrocephalus may develop due to its location (5). The CG is considered a glial neoplasm (6). CG is more commonly found in adult women, rarely in the pediatric age group (7-9). Our first case is more unusual since CG occurred in an 18 year-old woman. The CG mostly presents insidiously (74%) (10), rarely acutely (5). As described by Desouza and cols. (10) the CG mostly presents with headaches (40%) and visual defects (30%). Other symptoms described in these patients are memory deficits (24%), ataxia or incoordination (10%), endocrine disturbances with gonadotropic alterations as the most frequent (10%) (10). Most of the cases described are multisymptomatic (78%), being the most typical one, the presence of visual defects (10). In few patients, CG has been detected incidentally (3.7%) (1). In 2002 Oda and cols. (11) describe a case of a male with a two-year history of voracious appetite and progressive memory impairment three months prior to evaluation. Both of our cases presented severe headaches, with hypogonadism as the endocrine abnormality, and obstructive hydrocephalus in coincidence with the most common findings reported. CG should be included in the differential diagnosis of third ventricle’s masses. The most characteristic image suggestive of CG on MRI is an ovoid well-defined mass on the anterior region of the third ventricle or sellar region extending towards the hypothalamus. CG is usually isointense in T1 weighted images, with strong uniform contrast enhancement and with bilateral vasogenic edema (10). They may have cystic components (25%) and, though unusual, calcifications and hemorrhage can be found (10). On CT scans CG is hyperdense with homogeneous contrast enhancement. The lesion in our cases was typically oval, located in the suprasellar area, with a strong contrast enhancement on MRI, and hyperdense on CT in case 1. In case 1, the mass enhanced homogeneously after gadolinium 652

administration and was hyperintense on T2 weighted image, with third ventricle extension. Postoperatively, on T1 gadolinium weighted image the remnant tumor was inside the third ventricle with ventricular symmetry. In case 2, the suprasellar mass presented with heterogeneous enhancement after gadolinium administration with hypothalamic involvement and hydrocephalus on T1 weighted image, hyperintense on T2 weighted image. In the literature CG has been usually described as well circumscribed, ovoid in shape, isointense on T1 weighted MRI scans, with intense and uniform enhancement (12,13). Liu and cols. (14) found 63% of CG isointense on T1 weighted images, 70% with uniform enhancement and 42% hyperintense on T2 weighted images, aiding these findings in the differential diagnosis. The most difficult differential diagnosis is with craniopharyngiomas which have solid parts in adults. This was our presumptive diagnosis in case 2, inducing a transphenoidal approach. Tumors that originate in the anterior portion of the third ventricle should be considered. Pilocytic astrocytomas grow along optic structures and affect younger individuals, as it occurs in germinomas. Suprasellar meningiomas have the typical dural prolongation. Ependymomas have heterogeneous enhancement, frequent calcifications and internal hemorrhage (15). Histopathological findings consist of clusters and cords of epithelioid cells with abundant eosinophilic cytoplasm in a mucinous stroma with infiltrates of mature lymphocytes and plasma cells (2). By immunohistochemistry, they are positive for GFAP indicating the tumor’s glial origin (16). CG was confirmed in our cases due to the microscopic findings as well as the immunophenotype. TTF-1 transcription factor has been demonstrated to be constantly expressed in a series of 17 cases of CG of the third ventricle (17). Though not specific, this marker could help in diagnosis of this rare tumor. The treatment of choice is transcranial surgery, but incomplete resections are not uncommon. Mean time from onset of symptoms to surgical management has been described in 22.1 months (range 0-20 years) (18). In a review analyzing 54 operated patients, 22.7% recurred after partial resection and 74% died over a mean follow-up of 10.2 months (19). In a recent systematic review of 81 patients, the extent of surgical resection was reported in 75, of which 10.7% received Arch Endocrinol Metab. 2018;62/6


biopsy, 45.3% subtotal resection and 44% gross total resection (18). Ventricular valves are often needed due to obstructive hydrocephalus as in our two cases. Surgical approach has not been specified in many cases. In our first case the transcranial approach was chosen based on histopathologic possibilities obtained through the biopsy, since the best option for both diagnosis settled is the surgical resection through the transcranial approach. Our second case was approached endoscopally since was initially interpreted as a craniopharyngioma. There are no reports about the indication of extended endoscopic endonasal approach in gliomas, being our case the first described using this surgical approach. Currently, the most direct route to the suprasellar region is through an extended approach over the sella: the transplanum-transtuberculum approach, which allows the dissection of midline tumors over the pituitary gland and below the optic chiasm, without the need for brain retraction. This approach allows to follow the tumor mass located within the third ventricle and a visual control from below of the entire third ventricle, which cannot be obtained by any transcranial approach. Thus, the endoscopic approach has been used with more frequency in recent years for the treatment of tumors, especially craniopharingyomas, extending from the suprasellar region into the third ventricle (20). Surgical complications of endoscopic endonasal resection of tumors that extend into the third ventricle include neurological deficits as a result of direct neural tissue trauma or vascular compromise, pituitary hormonal dysfunction as a result of injury to the pituitary stalk or hypothalamus, and cognitive and psychological abnormalities as a result of injury to the frontal/ temporal lobes or hypothalamus. Cerebrospinal fluid leakage and sinonasal morbidity are the most common approach-specific complications (20). The most common postoperative complication described in the surgical treatment of CG is hypothalamic dysfunction, mostly diabetes insipidus and less frequently syndrome of inappropriate antidiuretic hormone (18). Short-term memory deficits are present in 8.8% (18). The translamina terminalis is associated with less postoperative morbidity, but without statistically significance (18). Post-operative complications in CG are DI (31%), amnesia (26%) and pulmonary embolism (15%) as the most frequent (10), with 38.3% of complications (18). Mortality in the immediate postoperative period is described in 32%, mainly due to thromboembolism, being higher after gross total resection. Ampie and Arch Endocrinol Metab. 2018;62/6

cols. (18) describe in a comprehensive review of 81 CG, 18 deaths (22.2%) at last follow up, mostly not related to disease progression, half of them within 18 days of surgery. Non-fatal postoperative complications are hypothalamic disorders and mental alterations (19). Gan and cols. (21) have recently described a high prevalence of obesity in a series of pediatric low grade gliomas affecting the optic pathway, hypothalamus and suprasellar areas (50% at 20 y). However, no statistical significant difference in the rate of overall complications has been shown between biopsy/ partial or gross resection (p = 0.82) (18). However, the most robust predictor of tumor control was gross total resection (18). In our first case surgery was performed with a partial resection, with few events on follow-up. In the immediate postoperative period, endocrine dysfunction was confirmed. Radiosurgery was indicated due to the remnant. Three years post treatment, the patient has only endocrine sequelae and no regrowth of the CG. The second case developed postoperatively infectious complications and finally died due to the presumptive diagnosis of thromboembolism. The high morbidity rate described in the surgical management of CG could be the reason for this fatal evolution, though more experience is needed to conclude about the benefits and risks of the endoscopic endonasal approach. Radiotherapy may be indicated when the resection is not complete. Progression or recurrence occurs in 30% of the patients; in these cases prognosis is usually bad. Nakajima and cols. (22) presented a case where the CG was resected and later treated by stereotactic radiosurgery. No regrowth was observed at 2-year follow-up. Kobayashi and cols. (19) described three cases of CG of the third ventricle with special emphasis on the effects of planned microsurgery and low dose stereotactic radiosurgery. However, other authors showed recurrences after radiotherapy (23,24). Our first case is another one where gamma knife has been used; 3 year-follow-up shows no recurrence with a stable remnant. However, the real efficacy of radiotherapy is still unclear. In these cases surgery was the main treatment by a transcranial or endonasal endoscopic approach, considering radiosurgery as an adjuvant therapy particularly in those cases of partial resections. Hypopituitarism is a plausible adverse event. However, CG might fatally evolve due to surgical complications. This postoperative mortality in the management of a low-grade tumor should be taken into consideration 653

CopyrightŠ AE&M all rights reserved.

Suprasellar chordoid glioma


Suprasellar chordoid glioma

in therapeutic decisions. Since the goal of treatment is complete total resection pursuing a curative intention, but due to the high perioperative morbidity, options should be balanced to define an individualized therapeutic approach. More experience is needed to bring conclusions about the endoscopic endonasal approach for the management of CG. Acknowledgments: nonthing to acknowledge. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Buccoliero AM, Caldarella A, Gallina P, Di Lorenzo N, Taddei A, Taddei GL. Chordoid glioma: clinicopathologic profile and differential diagnosis of an uncommon tumor. Arch Patol Lab Med. 2004;128:141-5. 2. Brat DJ, Scheithauer BW, Staugaitis SM, Cortez SC, Brecher K, Burger PC. Third ventricular chordoid glioma: a distinct clinicopathologic entity. J Neuropathol Exp Neurol. 1998;57(3):283-90. 3. Brat DJ, Scheithauer BW. Chordoid glioma of the third ventricle, in Louis DN, Ohgaki H, Wiestler OD, Cavanee WK (eds): WHO Classification of Tumours of the Central Nervous System, ed 4. Lyon, International Agency for Research on Cancer, 2007. 4. Cenacchi G, Rocaroli F, Cerasoli S, Ficarra G, Giangaspero F. Chordoid tumor of the 3d ventricle: immunohistochemical and ultrastructural study of 3 cases. Pathologica. 2000;92(2):132-3. 5. Carrasco R, Pascual JM, Reina T, Nieto S, Linera J, Sola RG. Chordoid glioma of the third ventricle attached to the optic chiasm. Successful removal through a trans-lamina terminalis approach. Clin Neurol Neurosurg. 2008;110(8):828-33. 6. Reifenberger G, Weber T, Weber RG, Wolter M, Brandis A, Kuchelmeister K, et al. Chordoid Glioma of the third ventricle: imminuhistochemical and molecular genetic characterization of a novel tumor entity. Brain Pathol. 1999;9(4):617-26. 7. Castellano-Sanchez AA, Schemankewitz E, Mazewski C, Brat DJ. Pediatric chordoid glioma with chondroid metaplasia. Pediatr Dev Pathol. 2001;4:564-67. 8. Goyal R, Vashishta RK, Singhi S, Gill M. Extraventricular unusual glioma in a child with extensive myxoid change resembling chordoid glioma. J Clin Pathol. 2007;60:1294-95. 9. Jain D, Sharma MC, Sarka C, Suri V, Rishi A, Garg A, et al. Chordoid Glioma: report of two rare examples with unusual features. Acta Neurochir (Wien). 2008;150:295-300.

12. Tonami H, Kamehiro M, Oquchi M, Higashi K, Yamamoto I, Njima T, et al. Chordoid glioma of the third ventricle: CT and MRI findings. J Comput Assist Tomogr. 2000;24:336-8. 13. Pasquier B, Péoc’h M, Morrison AL, Gay E, Pasquier D, Grand S, et al. Chordoid glioma of the third ventricle: a report of two new cases, with further evidence supporting an ependymal differentiation, and review of the literature. Am J Surg Pathol. 2002;26(10):1330-42. 14. Liu WP, Cheng JX, Yi XC, Zhen HN, Fei Z, Li Q, et al. Chordoid glioma: A case report and literature review. Neurologist. 2011;17:52-6. 15. Qixing F, Peiyi G, Kai W, Xuzhu Ch, Xiangde M, Jianping D. The radiological findings of chordoid glioma: report of two cases, one case with MR spectroscopy. Clinical Imaging. 2015;39:1086-9. 16. Wittchow R, Landas SK. Glial fibrillary acidic protein expression in pleomorphic adenoma, chordoma, and astrocytoma. A comparison of three antibodies. Arch Pathol Lab Med. 1991;115(10): 1030-3. 17. Bielle F, Villa Ch, Giry M, Bergemer-Fouquet AM, Polivka M, Vasiljevic A, et al. Chordoid Gliomas of the third ventricle share TTF-1 expression with organum vasculosum of the lamina terminalis. Am J Surg Pathol. 2015;39:948-56. 18. Ampie L, Choy W, Lamano JB, Kesavabhotla K, Mao Q, Parsa AT, et al. Prognostic factors for recurrence and complications in the surgical management of primary chordoid gliomas: A systematic review of literature. Clin Neurol Neurosurg. 2015;138:129-36. 19. Kobayashi T, Tsugawa T, Hashizume C, Arita N, Hatano H, Iwami K, et al. Therapeutic approach to chordoid glioma of the third ventricle. Three case reports and review of the literature. Neurol Med Chir (Tokyo). 2013;53:249-55. 20. Conger AR, Lucas J, Zada G, Schwartz TH, Cohen-Gadol AA. Endoscopic extended transsphenoidal resection of craniopharyngiomas: nuances of neurosurgical technique. Neurosurg Focus. 2014;37(4):E10. 21. Gan H-W, Phipps K, Aquilina K, Gaze MN, Hayward R, Spoudeas HA. Neuroendocrine morbidity after pediatric optic gliomas: a longitudinal analysis of 166 children over 30 years. J Clin Endocrinol Metab. 2015;100:3787-99. 22. Nakajima M, Nakasu S, Hatsuda N, Takeichi Y, Watanabe K, Matsuda M. Third ventricular chordoid glioma: case report and review of the literature. Surg Neurol. 2003;59(5):424-8. 23. Hanbali F, Fuller GN, Leeds NE, Sawaya R. Choroid plexus cyst and chordoid glioma. Report of two cases. Neurosurg Focus. 2001;10:E5. 24. Kurian KM, Summers DM, Statham PF, SmithC, Bell JE, Ironside JW. Third ventricular chordoid glioma: clinicopathological study of two cases with evidence for a poor clinical outcome despite low grade histological features. Neuropathol Appl Neurobiol. 2005;31:354-61.

Copyright© AE&M all rights reserved.

10. Desouza RM, Bodi L, Thomas N, Marsh H, Crocker M. Chordoid glioma: ten years of a low-grade tumor with high morbidity. Skull Base. 2010;20(2):125-38.

11. Oda M, Sasajima T, Kinouchi H, Sageshima M, Mizoi K. Third ventricular chordoid glioma: report of a surgical case. No Shinkei Geka. 2001;30(9):973-9.

654

Arch Endocrinol Metab. 2018;62/6


case report

Hypothyroidism associated with short bowel syndrome in children: a report of six cases Ananda Castro Vieira Passos1, Fábio de Barros1, Durval Damiani2, Beatriz Semer2, Wendy Cira Justiniano Cespedes2, Bruna Sannicola2, Ana Cristina Aoun Tannuri1, Uenis Tannuri1 Serviço de Cirurgia Pediátrica e Transplante Hepático, Laboratório de Investigação em Cirurgia Pediátrica (LIM-30), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil 2 Unidade de Endocrinologia Pediátrica, Divisão de Pediatria, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil 1

SUMMARY Short bowel syndrome (SBS) is the leading cause of intestinal failure in children, a condition of absence of sufficient bowel to meet the nutritional and metabolic needs of a growing individual. The treatment of patients in this situation is based on the association of parenteral and enteral nutrition for prolonged periods of time until intestinal rehabilitation occurs with complete enteral nutrition autonomy. Six consecutive cases of children with SBS (residual intestinal length of 5 cm to 75 cm) were managed with this program and were diagnosed with associated hypothyroidism during the treatment (ages at the diagnosis 5 months to 12 years). All patients were successfully treated with oral hormone reposition therapy and in one patient, the replacement was performed via rectal enemas due to a complete absence of small bowel. Although iodine deficiency associated to long-term parenteral nutrition is a well-known condition, this is the first report in the literature about an expressive number of patients with hypothyroidism detected in patients with SBS during the prolonged treatment for intestinal rehabilitation. Arch Endocrinol Metab. 2018;62(6):655-60

Correspondence to: Uenis Tannuri Faculdade de Medicina, Universidade de São Paulo Av. Dr. Arnaldo, 455, 4o andar, sala 4109 01246-903 – São Paulo, SP, Brasil uenist@usp.br Received on Oct/3/2018 Accepted on Oct/31/2018

INTRODUCTION

I

ntestinal failure in children occurs in the absence of sufficient bowel to meet the nutritional and metabolic needs of a growing and developing individual (1-4). Short bowel syndrome (SBS) is the leading cause of intestinal failure in children and is characterized by a small intestine length 25% shorter than expected for gestational age (5-7). The main causes of SBS in children are massive resections secondary to necrotizing enterocolitis, gastroschisis, intestinal atresia and intestinal volvulus (1-5,8). SBS-associated intestinal failure causes various metabolic changes in the body, many of which are associated with growth, intestinal adaptation and bone metabolic disease. Research on SBS and growth hormone (GH), glucagon-like peptide-2 (GLP-2), vitamin D and parathyroid hormone (PTH) is already well advanced; in contrast, little is known about the hypothalamic-pituitary-thyroid (HPT) axis and its balance in patients with SBS (3,8,9). Arch Endocrinol Metab. 2018;62/6

In critically ill patients with acute or chronic disease, decreased serum triiodothyronine (T3) and thyroxine (T4) levels are associated with normal or reduced concentrations of thyroid stimulating hormone (TSH). This set of findings is known as nonthyroidal illness syndrome (NTIS) (10-14). Periods of caloric restriction were also associated with low concentrations of T3 and T4 without increased TSH, suggesting that central action mechanisms are also responsible for inhibition of the HPT axis (11,12,15-17). It is not known whether these changes are part of a protective mechanism for decreasing basal metabolism and saving energy during critical periods or if they are part of an erroneous response to insult, motivated by the presence of inflammatory cytokines. Thus, accurate indications of the correction of these hormonal changes have not yet been fully established (10-12). In turn, the correct functioning of the HPT axis favors the intestinal adaptation process, and the replacement of thyroid hormones in experimental models of SBS has 655

Copyright© AE&M all rights reserved.

DOI: 10.20945/2359-3997000000093


Hypothyroidism and short bowel syndrome

shown benefits in intestinal adaptive parameters (i.e., enterocyte proliferation and increased crypt depth and villi height) (18). Finally, in addition to the thyroid changes caused by metabolic changes intrinsic to SBS, there is also the possibility of imbalance in the HPT axis secondary to iodine deficiency (19). HPT axis deficiency may lead to clinical hypothyroidism, which is classified as primary if the defect is at the thyroid gland, secondary if the problem is the TSH production by the hypophysis and tertiary if the hypothalamus does not produce thyrotropin releasing hormone (TRH) (13). The cases of six patients with SBS who presented changes in the HPT axis and hypothyroidism, during treatment of intestinal failure at the Children’s Institute of the University of São Paulo School of Medicine, from 2012 to 2017, are reported herein. As far as we know, this is the first report of an expressive number of patients with such association although iodine deficiency associated to long-term parenteral nutrition is a wellknown condition and may be avoided by adequate intravenous administration of sodium iodine (20).

REPORTS The medical records of 25 patients with short bowel syndrome treated at the Unit’s intestinal rehabilitation program were reviewed, of whom six presented with hypothyroidism (Table 1). It is important to note that all laboratory tests were repeated in order to adequately confirm the alterations.

currently been weaned from PN. Currently, the patient is at the 5th percentile for weight and height. Screening tests for thyroid function showed normal serum concentrations of TSH (3.95 μlU/mL; normal range 0.27 to 4.20 μlU/mL) and low serum concentrations of T3 (51 ng/dL; reference value [RV]: 80 to 200 ng/dL), T4 (4.7 µg/dL; RV: 5.1 to 14.1 µg/dL) and free T4 (0.73 ng/dL; RV: 0.93 to 1.70 ng/dL). In addition, pituitary and hypothalamic alterations were ruled out by laboratory tests (the serum levels of gonadotrophic hormones, growth hormone, adrenocorticotrophic hormone were normal, and magnetic resonance image was normal). After five months of hormone replacement therapy the levels of T3, T4 and free T4 (FT4) became normal, without significant clinical changes.

Case 2 PGFSV, 8 months old, SBS secondary to volvulus, with 35 cm of residual small bowel, dependent on PN since the time of intestinal resection performed at 3 months of age. Laboratory tests showed normal TSH (2.26 μlU/ mL; RV: 0.27 to 4.2 μlU/mL) and very low T4 and FT4 levels (3.4 μg/dL and < 0.3 ng/dL respectively). The laboratory tests and resonance image ruled out pituitary and hypothalamic alterations. At this time, the patient presented with severe malnutrition, with weight and height below the 5th percentile, hypoactivity and hyporeflexia. After 1 month of hormone replacement therapy, the patient’s T4 and FT4 levels returned to normal values, and the patient improved clinically, with weight gain and normal reactivity.

Case 1 RGPC, 4 years old, SBS secondary to gastroschisis and intestinal resection, with 75 cm residual small bowel, dependent on parenteral nutrition (PN) since birth, is

Case 3 PHLO, 4 years old, SBS secondary to gastroschisis, with 40 cm of residual small bowel, dependent on PN since

Table 1. Summary of clinical data of SBS patients with hypothyroidism Residual small bowel (cm)

Hypothyroidism cause

Nutritional status*

Daily calories Parenteral/ enteral (kcal/ kg/day)

Age at hypothyroidism

TSH** (μlU/mL)

T3/T4** (ng/dL)/ (µg/dL)

Free T4** (ng/dL)

1/new born

75

NTIS

Undernourished

70/75

3 years

3.9

51/4.7

0.7

Copyright© AE&M all rights reserved.

Patient/age at SBS

2/3 months

35

NTIS

Undernourished

40/130

7 months

2.3

- /3.4

< 0.3

3/new born

40

Iodine deficiency

Eutrophic

60/70

5 months

10.6

137/7.0

0.7

4/new born

50

Iodine deficiency

Eutrophic

- /200

3 years

19.8

110/8.1

1.5

5/8 years

15

Iodine deficiency

Eutrophic

50/ -

12 years

13.1

86/-

0.8

6/5 years

5

Iodine deficiency

Eutrophic

45/ -

11 years

> 300.0

< 47/< 0.8

< 0.3

* Nutritional status at the time of hypothyroidism diagnosis. ** Exams at the time of hypothyroidism diagnosis.

656

Arch Endocrinol Metab. 2018;62/6


Hypothyroidism and short bowel syndrome

Case 4 NVSM, 4 years old, SBS secondary to gastroschisis, with 50 cm of residual small bowel, PN dependent until 3 years of age, is not currently receiving PN, and he is maintaining stable nutritional status in the 10th percentile of weight and 2.5th percentile of height. Thyroid screening tests performed at 4 months of age showed no abnormalities. Subsequent tests at 3 years of age diagnosed primary hypothyroidism (TSH 19.79 μlU/mL, T3 110 ng/dL, T4 8.1 μg/dL, FT4 1.48 ng/ dL). Negative thyroid autoantibodies. The patient still receives hormone replacement therapy started at 3 years of age. Despite the improvement in laboratory tests, the administration of L-T4 did not cause significant clinical changes. Currently, the child feeds exclusively by oral route and has normal bowel movements.

Case 5 GHMM, 14 years old, SBS secondary to massive resection due to necrosis secondary to intestinal volvulus at 8 years of age. The patient currently has 15 cm of small intestine anastomosed to the transverse colon. The patient is dependent on PN daily and ingests minimal amounts of food orally. The patient remains eutrophic, in the 50th percentile of weight and height. At the age 12, during routine testing, the patient was diagnosed with primary hypothyroidism (TSH 13.14 µlU/mL, T3 86 ng/dL, 0.82). The thyroid autoantibodies were absent. The patient had no symptoms of hypothyroidism at the time of diagnosis. Oral administration of L-T4 normalized serum thyroid hormone concentrations although no evident clinical changes were noted. Arch Endocrinol Metab. 2018;62/6

Case 6 VTR, 11 years old, SBS caused by massive intestinal necrosis after bowel volvulus at 5 years of age. The patient has only part of duodenum, which is exteriorized via an ostomy in the mesogastrium, and 5 cm of rectum, which is buried in the pelvis. He was referred to our Service in bad conditions and undernourished (body weigh – 13 kg). The patient is totally PN-dependent and does not intake any food or medication orally. Despite that, he has been remained eutrophic, in the 50th percentile of weight and height. The clinical evaluations after 6 years of PN treatment showed excessive weight gain, hypoactivity, hyporeflexia and increased cervical thyroid gland volume (current body weight – 49kg). The laboratory tests showed that TSH was above 300 μlU/mL, T3 < 47 ng/dL, T4 < 0.8 μg/dL) and FT4 < 0.3 ng/dL. The thyroid autoantibodies were absent. Cervical ultrasound showed a diffusely enlarged thyroid with no nodules. Subsequent investigation indicated primary hypothyroidism due to iodine deficiency (due to problems involving pharmacological incompatibility with other trace elements, iodine was not included in PN solutions). L-T4 hormone replacement via rectal enemas normalized the patient’s thyroid hormone levels, and the patient presented significant clinical improvement, including weight loss. Finally, the data of these six patients were compared to the other 19 euthyroid patients with SBS cohort (Table 2).

DISCUSSION In the last century, there was only one reference in the literature reporting the occurrence of hypothyroidism in two pediatric patients with SBS under parenteral nutrition therapy (21). However, with the widespread prolonged use of this therapy in children and adults, few other publications have been showing such complication in the current century (22,23). We report here the occurrence of hypothyroidism observed in an expressive number of patients with SBS, during the period of intestinal rehabilitation. Considering the great number of patients with SBS, both children and adults, that have been submitted to prolonged periods of parenteral nutrition for the intestinal rehabilitation, we conclude the importance of the present publication. In fact, it is the sequence of a previous repor from our Service about the intestinal rehabilitation program for the treatment of children with SBS (20). 657

Copyright© AE&M all rights reserved.

the neonatal period until 2 years of age, is currently weaned from PN, and he is in the 50th percentile of weight and 25th percentile of height. The patient was diagnosed with primary hypothyroidism at 5 months of age (TSH 10.56 μlU/mL, T3 137 ng/dL, T4 7.0 μg/dL, FT4 0.73 ng/dL). The investigation for the presence of thyroid autoantibodies was negative. Administration of exogenous thyroid hormone for 1 year and 10 months promoted normalization of serum hormone concentrations. Subjectively, both the family and the healthcare team noticed a decrease in the volume and an increase in the consistency of bowel movements, with no other significant clinical changes after initiation of therapy. Currently, the patient does not receive levothyroxine (L-T4) and remains euthyroid.


Hypothyroidism and short bowel syndrome

Regarding iodine metabolism and parenteral nutrition therapy, an important historical aspect may be stressed. Since 20 or 25 years ago, the widespread practice of utilizing iodine-based antiseptics has been abandoned, although it prevented the patients to have iodine deficiency, even though this element was not administered. Certainly, the abandon of iodine-based formulas to 2% chlorhexidine solutions for skin and catheter hygiene may have caused iodine deficiency and hypothyroidism when iodine is not adequately provided to the patient (24). Similar to other centers, our parenteral nutrition solutions do not contain iodine, and supplies by oral route higher than the classical amount of 1 mg/kg/day may be considered in all children with any remaining intestine (25-28). Finally, in cases with relevant clinical symptoms of hypothyroidism, besides the iodine administration, the L-T4 hormone replacement may be advocated. The cases reported herein refer to children with the same underlying disease but with different hormonal axis changes. In the first two cases, the thyroid function findings were similar to those found in children with NTIS that is a condition more frequent than pituitary or hypothalamic related diseases. Also, it has already been described in patients with severe baseline pathologies (acute or chronic), and, to date, can be explained by increases in inflammatory cytokines, such as TNF-α, which block the activity of 5’-deiodinase (responsible for the conversion of T4 to T3) and other central mediators that reduce the release of TSH (10-13). Laboratory findings similar to those of NTIS are also observed in

experimental models of caloric restriction (acute or chronic) (12,15-17,29). There are no experimental studies that specifically relate SBS to the HPT axis; however, since SBS is a serious condition involving caloric restriction, we can infer that the mechanisms involved in thyroid changes in SBS are the same as those involved in NTIS and in experimental models of chronic caloric restriction. These two patients were treated with hormone replacement, and their T3, T4 and FT4 concentrations normalized. The first patient had no clinical changes after treatment and, despite having the longest residual bowel (75 cm), did not present significant intestinal adaptation and was still dependent on PN. The other patient presented significant clinical improvement, with reversal of hypoactivity and weight gain. However, this clinical response coincided with increased enteral and parenteral caloric intake and consequent improvement in the patient’s nutritional status. Therefore, it was difficult to conclude that hormone replacement was, in fact, beneficial or whether clinical improvement would occur regardless of L-T4 administration. Theoretically, the decrease in the basal metabolism caused by the downregulation of the HPT axis would be a protection mechanism during critical periods. In this sense, hormone replacement is debatable (10-17,29,30). Further studies are needed to define the actual role of L-T4 administration in these situations, especially in SBS, since thyroid hormones not only modulate basal metabolism but also have a trophic effect on the intestine, a fact that is especially important for children in the intestinal adaptation phase (18).

Table 2. Data comparing all SBS patients with respect to thyroid function and nutritional treatment Cases Age Age at SBS diagnosis

Iodine deficiency (cases 3, 4, 5, and 6)

Euthyroidism (19 patients)

8m – 4y

4y – 14y

1y – 7y

0m – 3m

0m – 8y

0m – 4m

Gastroschisis/volvulus

Gastroschisis/volvulus

Gastroschisis/volvulus/necrotizing enterocolitis/intestinal atresia

Residual small bowel (cm)

35 – 75

5 – 50

5 – 50

Age at hypothyroidism

7m – 3y

5m – 12y

No

Parenteral nutrition calories (%)

26 – 50

0 – 100

0 – 80

Undernourished

Eutrophic

Undernourished/Eutrophic

SBS cause

Nutritional status Iodine supplementation Copyright© AE&M all rights reserved.

Central hypothyroidism (NTIS – cases 1 and 2)

T3 (ng/dL) T4 (µg/dL) Free T4 (ng/dL) TSH (μlU/mL)

No

No

No

51.0

<47.0 – 137.0

82.0 – 179.0

3.4 – 4.7

<0.8 – 81.0

5.7 – 11.3

2.26 – 3.95/< 0.3 – 0.73

10.53 - >300 / <0.3 – 1.48

0.98 – 1.62

2.26 – 3.95

10.53 - >300

0.67 – 4.03

Normal Range: TSH 0.27 to 4.20 μlU/mL; T3 80 to 200 ng/dL; T4 5.1 to 14.1 µg/dL; Free T4 0.93 to 1.70 ng/dL.

658

Arch Endocrinol Metab. 2018;62/6


Hypothyroidism and short bowel syndrome

Arch Endocrinol Metab. 2018;62/6

have been few studies on the time needed to fully deplete iodine reserves, with some reporting that depletion would occur within a few months (19). Finally, one patient (case 6) presented an emblematic picture of clinical hypothyroidism secondary to a lack of iodine in parenteral nutrition administered solutions. This patient cannot intake no food orally, and even if he does, the whole food bolus would exit via the duodenostomy located 5 cm from the pylorus. The almost complete absence of intestinal absorption surface in this patient made it difficult to control their hypothyroidism. The administration of enteral iodine was not effective, and the administration of L-T4 orally also did not alter the serum concentrations of TSH and FT4. It is noticeable that the patient is currently receiving L-T4 rectally and has normal concentrations of thyroid hormones. This route of administration of the exogenous hormone has been previously described (33). In conclusion, changes in the HPT axis associated with SBS are multifactorial and involve other metabolic reactions that have not yet been fully elucidated. When the child becomes dependent on total parenteral nutrition and does not present conditions for oral food intake, iodine deficiency eventually leads to a primary hypothyroidism, condition similar to endemic goiter, and L-T4 replacement normalizes the clinical picture. In SBS cases where iodine deficiency is not as evident (i.e., partial enteral feeding), further studies are needed to define the complex relationships between SBS and the HPT axis and to guide therapy. It is important that the thyroid hormone levels be periodically evaluated in every child with SBS and included in a program of intestinal rehabilitation. Authorship: 1. Conception and design: Ananda Castro Vieira Passos, Fábio de Barros, Ana Cristina Aoun Tannuri, Uenis Tannuri. 2. Drafting the article and revising it critically: Durval Damiani, Beatriz Semer, Wendy Cira Justiniano Cespedes, Bruna Sannicola. 3. Final approval and all revisions of the version to be published: Uenis Tannuri. Disclosure: no potential conflict of interest relevant to this article was reported.

REFERENCES 1. Cole CR, Ziegler TR. Etiology and Epidemiology of Intestinal Failure. In: Duggan CP, Gura KM, Jaksic T (eds). Clinical Management of Intestinal Failure. CRC Press, Boca Raton, FL; 2012. p. 3-11. 2. Chakrabarty I, Burns DL. Pathophysiology of Intestinal Failure. In: Duggan CP, Gura KM, Jaksic T (eds). Clinical Management of Intestinal Failure. CRC Press, Boca Raton, FL; 2012. p. 13-30.

659

Copyright© AE&M all rights reserved.

The other four cases refer to patients with a diagnosis of primary hypothyroidism, with initial levels of TSH greater than 10 μlU/mL and low FT4 or FT4 at the lower limit of normal. The main causes of primary hypothyroidism are iodine deficiency, autoimmune thyroiditis, cervical radiotherapy and radioiodine therapy. These patients were negative for autoimmune thyroiditis, and none of them had undergone radiotherapy or radioiodine therapy. The cause of primary hypothyroidism in patients with SBS is likely multifactorial, and iodine deficiency (either due to low intake or non-absorption) plays a key role in this process. The iodine contained in ingested foods is absorbed in the small bowel (31) and is incorporated into the tyrosine residues of thyroglobulin molecules (13). It is known that minimal daily amounts of iodine (1 μg/kg) are sufficient to maintain the body’s reserve (32). Patients with SBS have insufficient small bowel length and do not absorb adequate amounts of micronutrients and should therefore receive them parenterally. It is interesting to comment that one of the patients (case 3), after achieving intestinal autonomy and becoming independent of PN, no longer needed hormone replacement. This case reinforces the idea of the relationship between intestinal failure and euthyroid status. In contrast, in another patient (case 4), this relationship was not evidenced, since despite the fact that the child is no longer dependent on PN to maintain stable growth and development, he still needs to receive L-T4. It is important to note that the other 19 children with SBS treated in our program of intestinal rehabilitation, with intestinal lengths similar to those reported herein, have no clinical hypothyroidism and have normal thyroid laboratory tests. The reason why children with similar intestinal lengths receiving similar amounts of nutritional and iodine intake manifest (or not) thyroid disease is not well established and requires further study. The last two cases are patients with SBS secondary to bowel volvulus. They fed normally until the age at which the volvulus occurred; therefore, they had normal iodine reserves. Both patients currently receive virtually no oral feeding and, consequently, negligible amounts of enteral iodine. Antisepsis of the devices of these patients is not performed with iodinated substances. One of them (case 6) developed clinical hypothyroidism 5 years after volvulus, and the other (case 5) was diagnosed with hypothyroidism (through laboratory investigation) 4 years after volvulus. There


Hypothyroidism and short bowel syndrome

3. Goulet O, Ruemmele F, Lacaille F, Colomb V. Irreversible intestinal failure. J Pediatr Gastroenterol Nutr. 2004;38:250-69. 4. Cooper A, Floyd TF, Ross AJ III, Bishop HC, Templeton JM Jr, Ziegler MM. Morbidity and mortality of short-bowel syndrome acquired in infancy: An update. J Pediatr Surg. 1984;19:711-8. 5. Galea MH, Holliday H, Carachi R, Kapila L. Short-bowel syndrome: A collective review. J Pediatr Surg. 1992;27:592-6. 6. Wales PW, Christison-Lagay ER. 2010 Short bowel syndrome: epidemiology and etiology. Semin Pediatr Surg. 2010;19:3-9. 7. Salvia G, Guarino A, Terrin G, Cascioli C, Paludetto R, Indrio F, et al. Neonatal onset intestinal failure: an Italian multicenter study. J Pediatr. 2008;153:674-6. 8. Carbonnel F, Cosnes J, Chevret S, Beaugerie L, Ngô Y, Malafosse M, et al. The role of anatomic factors in nutritional autonomy after extensive small bowel resection. JPEN J Parenter Enteral Nutr. 1996;20:275-80. 9. Abrams S. Osteopenia and Bone Health in Patients with Intestinal Failure. In: Duggan CP, Gura KM, Jaksic T (eds). Clinical Management of Intestinal Failure. CRC Press, Boca Raton, FL; 2012. p. 271-81. 10. Chopra IJ, Hershman JM, Pardridge WM, Nicoloff JT. Thyroid function in nonthyroidal illnesses. Ann Intern Med. 1983;6:946-57. 11. Economidou F, Douka E, Tzanela M, Nanas S, Kotanidou A. Thyroid function during critical illness. Hormones (Athens). 2011;10:117-24. 12. Bello G, Ceaichisciuc I, Silva S, Antonelli M. The role of thyroid dysfunction in the critically ill: a review of the literature. Minerva Anestesiol. 201076:919-28. 13. Menezes HC-Filho, Bedin MR, Manna TD Hipo e Hipertireoidismo. In: Damiani D (eds). Endocrinologia na Prática Pediátrica, 3th edition. Manole, Barueri, SP; 2016. p. 267-96. 14. Van den Berghe G. Non-thyroidal illness in the ICU: a syndrome with different faces. Thyroid. 2014;24:1456-65. 15. Vries EM, van Beeren HC, Ackermans MT, Kalsbeek A, Fliers E, Boelen A. Differential effects of fasting vs food restriction on liver thyroid hormone metabolism in male rats. J Endocrinol. 2015;224:25-35. 16. Shambaugh GE III, Wilber JF. The effect of caloric deprivation upon thyroid function-in the neonatal rat. Endocrinology. 1974;94:1145 (abstract). 17. van Haasteren GAC, Linkels E, van Toor H, Klootwijk W, Kaptein E, Jong FH, et al. Effects of long-term food reduction on the hypothalamus-pituitary-thyroid axis in male and female rats. J Endocrinol. 1996;150:169-78. 18. Yuksel O, Tatlicioglu E, Poyraz A, Sahin T, Pasaoglu H, Ekinci O, et al. Effects of thyroid hormone on the adaptation in short bowel syndrome. J Surg Res. 2009;155:116-24.

21. Puri P, Guiney EJ. Transient hypothyroidism associated with short gut syndrome. J Pediatr Surg. 1982;17:22-4. 22. Clarridge KE, Conway EE, Bucuvalas J. Hypothyroidism and iodine deficiency in an infant requiring total parenteral nutrition. JPEN. 2014;38:901-4. 23. Pearson S, Donnellan C, Turner L, Noble E, Seejore K, Murray RD. Endemic goiter and hypothyroidism in an adult female patient dependent on total parenteral nutrition. Endocrinol Diabetes Metab Case Rep. 2017;2017:1-3. 24. Moukarzel AA, Buchman AL, Salas JS, Vargas J, Guss W, Baron HI, et al. Iodine supplementation in children receiving long-term parenteral nutrition. J Pediatr. 1992;121(2):252-4. 25. Willard DL, Young LS, He X, Braverman LE, Pearce EN. Iodine content of enteral and parenteral nutrition solutions. Endocr Pract. 2017;23(7):775-9. 26. Belfort MB, Pearce EN, Braverman LE, He X, Brown RS. Low iodine content in the diets of hospitalized preterm infants. J Clin Endocrinol Metab. 2012;97:E632-6. 27. Ibrahim M, Morreale de Escobar G, Visser TJ, Durán S, van Toor H, Strachan J, et al. Iodine deficiency associated with parenteral nutrition in extreme preterm infants. Arch Dis Child Fetal Neonatal Ed. 2003;88:F56-7. 28. Cicalese MP, Bruzzese E, Guarino A, Spagnuolo MI. Requesting iodine supplementation in children on parenteral nutrition. Clin Nutr. 2009;28:256-9. 29. Araujo RL, Andrade BM, Figueiredo ASP, Silva ML, Marassi MP, Preira VS, et al. Low replacement doses of thyroxine during food restriction restores type 1 deiodinase activity in rats and promotes body protein loss. J Endocrinol. 2008;198:119-25. 30. Allan P, Lal S. Intestinal failure: a review. F1000Res. 2018;7:85. 31. Nicola JP, Basquin C, Portulano C, Reyna-Neyra A, Paroder M, Carrasco N. The Na+/I- symporter mediates active iodide uptake in the intestine. Am J Physiol Cell Physiol. 2009;296:C654-62. 32. Koletzko B, Goulet O, Hunt J, Krohn K, Shamir R. Guidelines on Paediatric Parenteral Nutrition of the European Society of Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) and the European Society for Clinical Nutrition and Metabolism (ESPEN), Supported by the European Society of Peadiatric Research (ESPR). J Pediatr Gastroenterol Nutr. 2005; 41 Suppl 2:S1-87. 33. Pinheiro CTC, Ybarra M, Franco RR, Setian N, Damiani D, Dichtchekenian V. Levotiroxina Retal para Tratamento de Hipotireoidismo Neonatal em Pacientes em Jejum Absoluto. 2015. Available at: http:// anais.sbp.com.br/trabalhos-de-congressos-da-sbp/11-congressobrasileiro-pediatrico-de-endocrinologia-e-metabologia/0134-levotiroxina-retal-para-tratamento-de-hipotireoidismo.pdf.

Copyright© AE&M all rights reserved.

19. Hardy G. Micronutrient Deficiencies in Intestinal Failure. In: Duggan CP, Gura KM, Jaksic T (eds). Clinical Management of Intestinal Failure. CRC Press, Boca Raton, FL; 2012.

20. Tannuri U, Barros F, Tannuri ACA. Treatment of short bowel syndrome in children. Value of the Intestinal Rehabilitation Program. Rev Ass Med Bras. 2016;62:575-83.

660

Arch Endocrinol Metab. 2018;62/6


acknowledgments As Editor-in-Chief, I would like to acknowledge and thank, also on behalf of the Associate Editors, all the reviewers who have dedicated their time to the AE&M.

Arch Endocrinol Metab. 2018;62/6

Debora Nazato Delaine Macedo Delmar Lourenço Denise Behegaray Denise Franco Denise Momesso Denise Pires de Carvalho Denise Zantut Wittmann Dhiãnah Chachamovitiz Domingos Malerbi Eduardo Luiz Wildemberg Elba Bandeira Elisa Lamback Enrrico Bloise Erico Carvalho Eugenia Rota Fabio Hecht Fabio Montenegro Fabio Moura Felipe Gaia Fernanda Andrade Filippo de Luca Flávia Conceição Flávia Lopes Flavia Valente Flavio Hojaij Francisco Finamor Fulvio Thomazelli Giovana Balarini Lima Glaucia Carneiro Gláucia Mazeto Hugo Monteiro Ibrahim Abdelrahman Abushouk Ihsan Ates Ivani Silva Izabella Costa Santos Jacqueline Rizzolli Janice Reis Jaqueline Horvath Joana Dantas João Roberto Martins Joaquim Silva Junior John Roger Andersen Jorge Pinto

Copyright© AE&M all rights reserved.

A Lori Brotto Adriane Cardoso Demartini Akihiro Nishimura Alexandre Hohl Alexandru Lonel Checherita Alexsandra Malaquias Alicia Motta Amanda Athayde Ana Hoff Ana Paula Fayh Andre da Silva Andre Lopes André Vianna Andreas Machens Angela Reichelt Antonio Ribeiro Oliveira Jr Arnaldo Bordon Bianca Almeida Pititto Bibiana Colenci Bo Ban Bruno Halpern Cacio Ricardo Carlos Antonio Negrato Carlos Penatti Carolina Cunha de Oliveira Carolina Ferraz Carolina Janovsky Cecilia Helena Gouveia Célia Regina Nogueira Charles Marques Lourenço Cibele Borges Cintia Marques Clarissa Martins Claudia Maria Cenci Claudia Ridel Juzwiak Cleber Camacho Cléo Mesa Junior Crésio Alves Cristiane kochi Cynthia Valerio Daniel Bulzico Daniel Kendler Danilo Villagelin Debora Bohnen

661


Copyright© AE&M all rights reserved.

José Miguel Dora José Sgarbi Kelly Giudici Konstantinos Tziomalos Kuanfeng Xu Larissa Gomes Leila de Paula Leila Nikniaz Lenita Zajdenverg Licio Velloso Lucas Cunha Luciana Bahia Luciana Pechmann Luciana Viana Lucio Vilar Luiz Kraemer Aguiar Malachi Mckenna Manoel Martins Marcello Bertoluci Marcio Krakauer Marcio Machado Marcio Mancini Marcus Leitão Marcus Leitão Margaret Boguzewski Maria Aparecida koike Folgueira Maria Candida Fragoso Maria Caroline Coelho Maria Edna Melo Maria Izabel Chiamolera Maria Lazo Porras Maria Lucia Farias Maria Tereza Nunes Marina kizys Mario Campos Mário Lúcio Araújo Jr Mauro Czepielewski Melissa Preamor Milena Caldato Mimi Kim Monica Gabbay Monique Ohe Nágila Damasceno Nara Crispim Narriane Chaves Nelson Rassi Nelson Wohlk Nina Musolino

662

Patricia Dualib Patricia Texeira Paula Elias Paulo Alonso Alves Junior Pedro Rosario Pierpaolo Trimboli@eoc.ch Rachel Honjo Rafael Machry Rafael Scheffel Ralph Brinks Ramon Ramos Raquel Jallad Renan Montenegro Jr Renata Leborato Ricardo Delfim Ricardo Oliveira Robert C Hood Roberta Tarantino Roberto Luis Zagury Rodrigo Moreira Rosana Pereira Marques Rosita Fontes Ruth Mangupli Sandra Benite Sandra Ferreira Sandro Sperandei Sergio Dib Sergio Vencio Sharon Parish Sheila Lecke Shirlei de Súss Silmara Leite Dalva Poyares Solange Travassos Suemi Marui Susan Lindsey Sylka Rodovalho Taís Assmann Taíse Cerqueira Tania Carvalho Ortiga Valdir Moises Vanda Jorgetti Vania Nunes Victória Borba Wenjun Cao Yukiko Hayashi Zeynep Silklar Zuleica Fortes

Arch Endocrinol Metab. 2018;62/6


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.