0
Thank you to our sponsors!
1
IMF Patient and Family Webinar Agenda
*times listed in US Eastern Time Zone
10:00 – 10:05 AM 10:05 – 10:25 AM
Welcome Announcements with Dr. Brian G.M. Durie Myeloma 101 Dr. Brian G.M. Durie
10:25 – 10:50 AM
Life Is A Canvas, You Are The Artist: Side Effects & Symptom Management
Teresa Miceli RN, BSN, OCN 10:50 – 11:10 AM
Addresses in Myeloma Treatments: Looking Beyond 2021 Dr. Brian G.M. Durie
11:10 – 11:20 AM
Panel Discussion
11:20 – 11:30 AM
BREAK
2
IMF Patient and Family Webinar Agenda
*times listed in US Eastern Time Zone
11:30 – 12:00 PM
Immune Therapies: Treatments That Work With Our Own Immune Systems Dr. Adam Cohen
12:00 – 12:20 PM
How to Find What You Need to Know Robin Tuohy, IMF Vice President, Support Groups
12:20 – 12:50PM
When Myeloma Comes Back: Approaches to Relapse Dr. Rafat Abonour
12:50 – 1:00 PM
Summary Panel Discussion Webinar Survey & Closing Remarks 3
IMF Patient and Family Webinar
Myeloma 101 and COVID-19 updates Brian G.M. Durie, MD Cedars-Sinai Outpatient Cancer Center Los Angeles, CA 4
Global cases of COVID-19 reported
Data from the CDC and WHO. The data includes cases as of October 25, 2021. https://covid.cdc.gov/covid-data-tracker/#global-counts-rates and https://covid19.who.int/
5
Overall U.S. COVID-19 Data Tracker Trends in Number of COVID-19 Cases and Deaths in the US Reported to CDC
Overall US COVID-19 Vaccine | Deliveries and Administration; Maps, charts, and data provided by CDC https://covid.cdc.gov/covid-data-tracker/#vaccinations and https://covid.cdc.gov/covid-data-tracker/#datatracker-home
6
Delta Variant: Infections and Spread
CDC -- Delta Variant: What We Know About the Science https://www.cdc.gov/coronavirus/2019-ncov/variants/delta-variant.html
7
Vaccination for Myeloma Patients YES is good!!! Checking antibody levels becoming more important Additional vaccine dose strongly recommended 4th dose (booster) under review Please note: Antibody tests for COVID-19 are available through healthcare professionals and laboratories. Check with your healthcare professional to see if they offer antibody tests and whether you should get one. https://www.cdc.gov/coronavirus/2019-ncov/testing/serology-overview.html
8
Which vaccine? Pfizer/ Moderna – Excellent!
J&J – not live!/ single shot only Blood clot risk AstraZeneca – 79%76% efficacy
(slightly reduced) Blood clot risk
100% reduction of hospitalizations and deaths
9
Important to know Airborne transmission dominant Variants a serious challenge Beware asymptomatic spread Watch out for young people! Still avoid travel Indoor space a severe problem 10
Risks of Air Travel
Significant Beware the food service time (maybe skip) Boarding/ disembarking a concern One infected individual can infect plane
Still considered as essential only!
11
Masks – still required! So: use best masks possible – BROAD AirPro Mask AirPro Mask is a Powered Air-Purifying Respirator system. It includes a KN95 mask and a filtration unit with a 3-speed fan, HEPA H13 filter, and chargeable lithium battery.
CDC: Your Guide to Masks https://www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/about-face-coverings.html
12
What Can You Do Once You’re Vaccinated?
New York Times – March 30, 2021 https://www.nytimes.com/interactive/2021/03/30/opinion/coronavirus-vaccine-risks.html
13
When you’ve been fully vaccinated
CDC: When You’ve Been Fully Vaccinated - How to Protect Yourself and Others https://www.cdc.gov/coronavirus/2019-ncov/vaccines/fully-vaccinated.html
14
Patient Guidance Question: Can I go for a walk?
• Yes Question: Can I go to the dentist?
• Yes, with care 15
Common Issues Pandemic fatigue/ stress Not feeling resilient Healthcare team not coordinated Not prepared for Zoom or appointments Need electronic help! Contacts with unknown COVID-19 status 16
Pandemic Fatigue Real for everyone!
Important to reach out to family and friends Try to de-stress
Shedd Aquarium – Penguins Work on resilience Living Well with Myeloma: Building Your
Resilience During Challenging Times (IMF video) Resilience Builder Tool (link on IMF website)
17
Who is eligible for a COVID-19 Vaccine Booster Shot?
CDC: Who Is Eligible for a COVID-19 Vaccine Booster Shot? https://www.cdc.gov/coronavirus/2019-ncov/vaccines/booster-shot.html
18 18
Vaccination Boosters and Antibody Levels
Current questions
19
COVID-19 vaccination in patients with multiple myeloma: a consensus of the European Myeloma Network
COVID-19 vaccination in patients with multiple myeloma: a consensus of the EMN https://www.thelancet.com/journals/lanhae/article/PIIS2352-3026(21)00278-7/fulltext
20 20
What does the future hold?
Vaccination essential.
Masks required. Caution in indoor spaces.
21
Other IMF Resources: Ask Dr. Durie Videos
COVID-19 FAQ #35: What should myeloma patients know about antibody testing and booster shots?
COVID-19 FAQ #34: How can myeloma patients protect themselves
https://www.myeloma.org/videos/covid-19-faq-35-shouldmyeloma-patients-know-about-antibody-testing-booster-shots
https://www.myeloma.org/videos/covid-19-faq-34-how-canmyeloma-patients-protect-themselves-against-delta-variant
against the Delta variant?
22
Support Group Virtual Meetings
Over 90 support groups are now holding monthly virtual GoToMeetings through the IMF
23
Support Group Virtual Meetings
Over 90 support groups are now holding monthly virtual GoToMeetings through the IMF
24
We will get through this together! Myeloma has no borders
“Do Remember They Can’t Cancel the Spring” – David Hockney
Support messages in the sky above Los Angeles An apricot tree grows in Turkey
25
IMF Patient and Family Webinar
Teresa Miceli RN, BSN, OCN Mayo Clinic Rochester, MN
Life Is A Canvas, You Are The Artist: Side Effects & Symptom Management 26
Patient Family Seminar, October 30, 2021
LIFE IS A CANVAS, YOU ARE THE ARTIST Name Teresa Miceli, RN BSN OCN Institution Mayo Clinic-Rochester, MN
Patient Education Slides 2021
OBJECTIVES FRAMING YOUR CARE
Know your care team, Shared Decision Making & Reliable Resources
COLOR WHEEL OF TREATMENT
Myeloma and treatment side effects & symptom management
LIVE LIFE IN COLOR
Healthful Living, infection prevention, Learn Lots, Laugh renal and bone Much, health Love Life 28
FRAMING YOUR CARE Know your care team, Shared Decision Making & Reliable Resources
You are central to the care team
Pharmacist
CARE TEAM COLLAGE General Hem/Onc Myeloma Specialist
Communicate with your team • Myeloma is a chronic disease • Understand the roles of each
team member and who to contact for your needs • Participate in support network
Be empowered • Ask questions, learn more • Participate in decisions • Know your history –
Myeloma ManagerTM Personal Care AssistantTM
Primary Care Provider (PCP)
You and Your Caregiver(s) Support Network
Subspecialists Allied Health Staff
30
SHARED DECISION-MAKING Be empowered to be part of the treatment decision-making • Ask for time to consider options (if needed/appropriate) • Understand options; consider priorities • Use reliable sources of information • Use caution considering stories of personal
experiences • Consider your goals/values/preferences • Get a 2nd opinion, Meet with a Myeloma expert
Data From Research
HCP Clinical Experience TREATMENT DECISION
• Express your goals/values/preferences; create a
dialog
• My top priority is [goal/value]; additional [preferences] are also important. • I think [treatment] may be a good choice given my priorities… What do you think?
• Arrive at a treatment decision together
Your Preference Philippe Moreau. ASH 2015.
31
KNOWLEDGE IS POWER USE REPUTABLE SOURCES Website: http://myeloma.org
IMF TV Teleconferences
eNewsletter: Myeloma Minute
IMF InfoLine 1-800-452CURE 9am to 4pm PST Download or order at myeloma.org
32
COLOR WHEEL OF TREATMENT Treatment options, side effects, symptom management, & supportive care
GALLERY OF GOALS MYELOMA TREATMENT • Rapid and effective disease control • Durable disease control • Improved overall survival • Minimize side effects
SUPPORTIVE THERAPIES • Prevent disease- and treatmentrelated side effects • Optimize symptom management • Allow for good quality of life
• Allow for good quality of life
DISCUSS GOALS AND PRIORITIES WITH YOUR HEALTHCARE TEAM 34
PATIENT-REPORTED SYMPTOMS A meta-analysis identified the most common patient-reported symptoms and impact on QOL and were present at all stages of the disease. Symptoms resulted from both myeloma disease and treatment, including transplant, and were in these categories:
Physical
Psychological
• Fatigue
• Depression
• Constipation
• Anxiety
• Pain
• Sleep Disturbance
• Neuropathy
• Decreased Cognitive Function
• Impaired Physical Functioning • Sexual Dysfunction
Ramsenthaler, et al. 2016. https://doi.org/10.1111/ejh.12790.
Financial • Financial burden (80%) • Financial toxicity (43%)
• Decreased Role & Social Function
35
COLOR WHEEL OF TREATMENT OPTIONS -Mibs
Frontline
Velcade® (bortezomib)
Maintenance
Velcade® (bortezomib)
Relapse
Kyprolis® (carfilzomib) Ninlaro® (ixazomib)
-MAbs
-Mides
Steroids
Darzalex® (daratumumab)
Thalomid® (thalidomide) Revlimid® (lenalidomide)
Dexamethasone Prednisone Prednisolone SoluMedrol
Others
Melphalan Cyclophosphamide
Darzalex® (daratumumab) Empliciti® (elotuzumab) Sarclissa® (Isatuximab)
Thalomid® (thalidomide) Revlimid® (lenalidomide) Pomalyst® (pomalidomide)
Dexamethasone Prednisone Prednisolone SoluMedrol
Melphalan Cyclophosphamide Bendamustine
CelMods • Iberdomide • CC-92480
Neuropathy Carfilzomib: Cardiac
ImmunoTherapy
Cellular Therapies Melphalan + ASCT
Revlimid® (lenalidomide)
Pending FDA Approval Noted Side effects
Alkylators
Infusion reaction
DVT/PE
See steroid slide
Myelosuppression
Blenrep® (Belantamab mafodotin) “Belamaf”
Xpovio® (Selinexor) Doxil (liposomal doxorubicin) Farydak® (panobinostat)
Abecma® (Ide-Cel, CAR-T)
ADCs BSAs Ex: Teclistamab, Talquetamab Cevostamab
Venclexta® (venetoclax)
Other CAR-T • Cilta-Cel
Infusion reaction Blenrep®: Keratopathy
Myelosuppression, GI Selinexor: Low sodium
Infection risk CAR-T: CRS and neurotoxicity
DVT – Deep Vein Thrombosis; PE – Pulmonary Embolism; ADC – Antibody Drug Conjugates; BSA – Bi-Specific Antibodies; ASCT – Auto Stem Cell Transplant
Melphalan + ASCT
36
THE BRIGHT
&
DARK SIDE TO STEROIDS
Steroid Synergy Steroids are a backbone and work in combination to enhance myeloma therapy
Managing Steroid Side Effects • Consistent schedule (AM vs. PM) • Take with food • Stomach discomfort: Over-the-counter or
prescription medications • Medications to prevent shingles, thrush, or other infections
Do not stop or adjust steroid doses without discussing it with your health care provider
Steroid Side Effects • Irritability, mood swings, depression • Difficulty sleeping
• Blurred vision, cataracts
(insomnia), fatigue
• Flushing/sweating
• Increased risk of
• Stomach bloating,
infections, heart disease
• Muscle weakness,
cramping
• Increase in blood
pressure, water retention
hiccups, heartburn, ulcers, or gas
• Weight gain, hair
thinning/loss, skin rashes
• Increase in blood
sugar levels, diabetes
Rajkumar SV, Jacobus S, Callander NS, Fonseca R, Vesole DH, Williams ME, Abonour R, Siegel DS, Katz M, Greipp PR, Eastern Cooperative Oncology Group (2010) Lenalidomide plus high-dose dexamethasone versus lenalidomide plus low-dose dexamethasone as initial therapy for newly diagnosed multiple myeloma: an open-label randomised controlled trial. Lancet Oncol 11(1):29–37. King T, Faiman B. Steroid-Associated Side Effects: A Symptom Management Update on Multiple Myeloma Treatment. Clin J Oncol Nurs. 2017 Apr 1;21(2):240-249. doi: 10.1188/17.CJON.240-249. PMID: 28315528.
37
HIGHLIGHT YOUR CARE:
ADDITIONAL TOOLS TO COMPLETE THE PICTURE DVT/PE Prevention
Medications
Non-medication Therapies
Lifestyle Options
Bone Health
Blood thinners Ex: Aspirin, DOACs
Bone Strengthening Agents Calcium Vitamin D
Compression stockings
Radiation Surgery Immobilization Physical therapy
Activity Stop smoking Weight loss
Activity
Renal Health
Infection Prevention
Peripheral Neuropathy
GI Symptoms
Med dose reduction Avoid harmful meds
Antibacterial Antiviral Antifungal IVIG GCSF
Antidepressants Anti-neuroleptic Analgesia Vitamins Dose adjustments
Anti-nausea Anti-diarrheal Laxatives & stool softeners Fiber-binding agents
Dialysis
Masking Activity
Massage Acupuncture Cocoa Butter
Dietary choices Relaxation
Hydration
Handwashing Avoid crowds & sick people Monitor for fever COVID precautions
Activity Diabetes management
Avoid greasy foods Activity Hydration
38
GI SYMPTOMS: PREVENTION & MANAGEMENT Diarrhea may be caused by medications and supplements • • • •
Laxatives, antacids with magnesium Antibiotics, antidepressants, others Milk thistle, aloe, cayenne, saw palmetto, ginseng Sugar substitutes in sugar free gum
Avoid caffeinated, carbonated, or heavily sugared beverages Take anti-diarrheal medication • Imodium®, Lomotil®, or Colestid if recommended • Fiber binding agents – Metamucil®, Citrucel®, Benefiber® • Welchol® if recommended
Physical
Constipation may be caused by • Opioid pain relievers, antidepressants, heart or blood pressure medications, others • Supplements: Calcium, Iron, vitamin D (rarely), vitamin B-12 deficiency
Increase fiber • Fruits, vegetables, high fiber whole grain foods • Fiber binding agents – Metamucil®, Citrucel®, Benefiber®
Fluid intake can help with both diarrhea and constipation, and good for kidneys. Discuss GI issues with health care providers to identify causes and make adjustments to medications and supplements.
Smith LC, et al. CJON.2008;12(3)suppl:37-52. Faiman B. CJON. 2016;20(4):E100-E105. Miceli, T. et al. (2021). Cancer Treatment and Res Com, 100476.
39
PAIN: PREVENTION AND MANAGEMENT
Physical
Pain can significantly compromise quality of life Sources of pain include bone disease, neuropathy and medical procedures
Management • Prevent pain when possible • Bone strengtheners to decrease fracture risk; anti viral to prevent shingles; sedation before procedures
• Interventions depends on source of pain • May include medications, physical therapy, surgical intervention, radiation therapy, etc
• Complementary therapies • Mind-body, meditation, yoga, supplements, acupuncture, activity, etc
Faiman B, et al. CJON. 2017;21(5)suppl:19-36. Miceli, T. et al. (2021). Cancer Treatment and Res Com, 100476.
Tell your health care provider about any new bone pain or chronic pain that is not adequately controlled
40
PERIPHERAL NEUROPATHY MANAGEMENT Peripheral neuropathy: damage to nerves in extremities (hands, feet, or limbs) • • • • • •
Numbness Tingling Prickling sensations Sensitivity to touch Burning and/or cold sensation Muscle weakness
Physical
Prevention / Management: • Bortezomib once-weekly or subcutaneous administration • Massage area with cocoa butter regularly • Supplements:
• B-complex vitamins (B1, B6, B12) • Folic acid, and/or amino acids but do not take on day of Velcade® (bortezomib) infusion
• Safe environment: rugs, furnishings, shoes
If PN worsens, your HCP may: Report symptoms of peripheral neuropathy early to your health care provider; nerve damage from PN can be permanent if unaddressed
• Change your treatment • Prescribe oral or topical pain medication • Suggest physical therapy
Faiman B, et al. CJON. 2017;21(5)suppl:19-36. Tariman, et al. CJON.2008;12(3)suppl:29-36. Miceli, T. et al. (2021). Cancer Treatment and Res Com, 100476.
41
FATIGUE, ANXIETY & DEPRESSION
Physical Psychological
Physical symptoms impact mental well being. All can affect quality of life and relationships.
• Fatigue is the most common reported symptom (98.8%) Sources include anemia, pain, reduced activity, insomnia, treatment toxicity, bone marrow suppression
• Anxiety reported in >35% • Depression nearly 25% Financial concerns, disease progression, end-of-life, and change in social and sexual function were highlighted sources
Often, people do not share these symptoms with their provider. Talk to your provider about symptoms that are not well controlled or thoughts of self harm. Help is available. Ramsenthaler, et al. 2016. https://doi.org/10.1111/ejh.12790. Catamero D et al. CJON. 2017; 21(5)suppl:7-18.
42
REST AND RELAXATION CONTRIBUTE TO GOOD HEALTH � Adequate rest and sleep are
essential to a healthful lifestyle
Short and disturbed sleep increase risk of • Heart related death • Increase anxiety • Weaken immune system • Worsened pain • Falls and personal injury
� Things that can interfere with sleep • Medications : steroids, stimulants, herbal
supplements, alcohol • Psychologic: fear, anxiety, stress • Physiologic: sleep apnea, nocturia, pain, inactivity, heart issues Rod NH et al 2014. PloS one. 9(4):e91965; Coleman et al. 2011. Cancer Nurs. 34(3):219-227. National Sleep Foundation. At: http://sleepfoundation.org/ask-the-expert/sleep-hygiene
Psychological
� Sleep hygiene is necessary for
quality nighttime sleep, daytime alertness
• Engage in exercise but not too near • • • • • •
bedtime Increase daytime natural light exposure Avoid Daytime napping Establish a bedtime routine - warm bath, cup of warm milk or tea Associate your bed ONLY with sleep Sleep aid may be needed Avoid before bedtime: • Caffeine, nicotine , alcohol and sugar • Large meals and especially spicy, greasy foods • Computer screen time
Mustian et al. Journal of clinical Oncology. Sep 10 2013;31(26):3233-3241. Stan DL, et al. Clin J Oncol Nurs. Apr 2012;16(2):131-141. Zeng Y et al., Complementary therapies in medicine. Feb 2014;22(1):173-186.
43
HEALTHFUL LIVING STRATEGIES: PREVENTION Manage stress • Rest, relaxation, sleep hygiene • Mental health / social engagement • Complementary therapy
Maintain a healthy weight • Nutrition • Activity / exercise
Preventative health care • Health screenings, vaccinations • Prevent falls, injury, infection • Stop smoking
Maintain renal health • Myeloma management • Hydration • Avoid renally-toxic medications – Dose adjust to renal function • Diabetes management
Protect your bones • Nutrition, Calcium + D supplement • Weight-bearing activity / walking • Bone strengthening agents
• Dental care
“An ounce of prevention is worth a pound of cure.” Benjamin Franklin Faiman B, et al. CJON. 2017;21(5)suppl:19-36. Dimopoulous M, et al. Leukemia. 2009;23(9):1545-56. Brigle K, et al. CJON. 2017;21(5)suppl:60-76. Faiman B, et al. CJON. 2017;21(5)suppl:19-36. Faiman B, et al. CJON. 2011;15suppl:66-76. Miceli TS, et al. CJON. 2011;15(4)suppl:9-23.
44
Financial
FINANCIAL BURDEN Financial burden comes from
Funding and assistance may be available
• Medical costs
• Federal programs
• • • •
Premiums Co-payments Travel expenses Medical supplies
• Prescription costs
• Loss of income • Time off work or loss of employment • Caregiver time off work
Contact the Social Services department at your hospital or clinic to talk to a social worker for assistance.
• Pharmaceutical support
• Non-profit organizations • Websites: • • • • • • •
Medicare.gov SSA.gov LLS.org Rxassist.org NeedyMeds.com HealthWellFoundation.org Company-specific website
45
YOU ARE NOT ALONE
Questions?
IMF Patient and Family Webinar
Advances in Myeloma Treatments: Looking Beyond 2021 Brian G.M. Durie, MD Cedars-Sinai Outpatient Cancer Center Los Angeles, CA 47
Advances in Myeloma Treatments: Looking Beyond 2021
DR. BRIAN DURIE
Brian GM Durie Patient and Family Webinar
Progress in All Areas Diagnosis, risk criteria and monitoring Early disease
Frontline therapy Consolidation/ maintenance Early relapse Later relapse and new drug development 49
MGUS Early detection (from screening) will: Reveal possible causes or triggers
Help improve all outcomes Enable early interventions for both prevention and treatment 50
iStopMM Project Details Ca 3600 with MGUS Smoldering myeloma N=200
No further work-up N=1200 •
• • • •
Without MGUS/SMM
R IMWG Reccommendations N=1200
Intervention arm N=1200
Followed for progression to multiple myeloma or another malignancy (Cancer Registry) Followed for all diagnoses in inpatient and outpatient clinics throughout Iceland (Patient Registry) Followed for vital status every 2 months (Population Registry) All prescriptions throughout Iceland (Prescription registry) Regular quality of life assessments 51
A Glimpse of the Origins of MGUS 12/1997 No MGUS detected
MGUS – 8/2006
Mass spec positive 24032.4 24032.6
Dr. Kyle – Olmsted County MGUS Study
52
Role of biobanking in iStopMM Material biobanked Bone marrow • BM biopsy -> formalin fixed
• BM smear • BM aspirate -> plasma + cell sorting Blood and urine • LiHep plasma
• • • • • • •
Cell free Plasma Buffy coat EDTA tube (DeCode) Serum Mononuclear cells PAXgene tube Urine 53
Many diverse outcomes of iStopMM Tumor microenvironment Cancer screeningRenal amyloidosis Risk scores MGRS Mental health Pain Genetics Early intervention Flow cytometry Neuropathy Correlative science Waldenströms Survival Cardiac amyloidosis Psychological impact Polyclonal The significance of the immunoglobulins “unknown significance”
Smoldering Multiple Myeloma (SMM) SMM risk scoring is essential Better predictors are coming! Best management is evolving
55
Risk Score to Predict Progression Risk At 2 Years Score
100 High-risk group >12
0 2 3 5
0 3 4 0 2 3 5 6 2
80
% with progression
Risk Factor FLC Ratio 0-10 (ref) > 10-25 > 25-40 > 40 M protein (g/dL) 0-1.5 (ref) > 1.5-3 >3 BMPC% 0-15 (ref) > 15-20 > 20-30 > 30-40 > 40 FISH abnormality
Intermediate-risk group (9-12)
Risk Stratification Groups
Hazard Ratio (95% CI) Versus Low-risk group (censored 2 year)
0-4
Reference
5-8
7.56 (3.77 to 15.2)
9-12
17.3 (8.63 to 34.8)
> 12
31.9 (15.4 to 66.3)
Low-intermediate-risk Group (5-8)
60
40 Low-risk group (0-4) 20
Total Risk Score
2-year Progression n (%)
0-4
9 / 241 (3.7%)
5-8
67 / 264 (25.4%)
9-12
65 / 133 (48.9%)
> 12
37 / 51 (72.6%)
0 0
6
12
18
24
30
36
42
48
54
60
Months # at Risk 0-4
241
238
229
213
194
175
153
117
100
76
63
5-8
264
256
229
197
174
145
118
91
73
53
44
9-12
133
119
98
73
59
47
33
26
20
14
13
>12
51
41
29
21
14
9
7
5
2
2
2
San Miguel. ASCO 2019. Abstr 8000. Mateos Blood Cancer J. 2020;10:102.
Ultra High Risk
56
Curative Strategy for High-Risk Smoldering Myeloma (GEM-CESAR) Outcomes including Consolidation & 1 year maintenance 77 patients completed induction, HDT-ASCT, consolidation, and 1 yr of maintenance Induction (KRd x 6) (n = 77)
HDT-ASCT (n = 77)
Consolidation (KRd x 2) (n = 77)
Maintenance (Rd x 1 Yr) (n = 77)
≥ CR
43
63
75
81
VGPR
43
24
18
13
PR
13
13
7
5
Progressive disease
--
--
--
1*
MRD negative
33
49
65
62
Response, %
≥ CR 81%
MRD 62%
*Biological progressive disease at end of maintenance, MRD positive.
Mateos. ASH 2019. Abstr 781.
57
Curative Strategy for High-Risk Smoldering Myeloma (GEM-CESAR): PFS and OS 100
100
PFS 35-Mo PFS: 92%
60 40 20 0
OS
80
Patients (%)
Patients (%)
80
35-Mo OS: 96%
60 40 20
Median follow-up: 35.2 (5.4-53.2) 0
10
20
0 30
40
50
Mos
6 patients progressed (biological PD, n = 5) ‒ 4 patients with PD were at ultrahigh risk Mateos. ASH 2019. Abstr 781.
Median follow-up: 35.2 (5.4-53.2) Median follow-up: 35.2 (5.4-53.2) 0
10
20
30
40
50
60
Mos
3 patients died; only 1 was considered a treatment-related death 58
Blood Testing for Monitoring Mass spec gives more precision Blood myeloma cells can be tracked Immune status can also be monitored Interim MRD status can be assessed (reducing need for bone marrows) 59
Evidence for role of blood testing
Circulating Tumor Plasma Cell (CTPC) Black Swan Research Initiative Project Publications
Link to articles -- https://pubmed.ncbi.nlm.nih.gov/30455467/ and https://pubmed.ncbi.nlm.nih.gov/31697808/
60
MRD status systematically discriminates two different risk groups A large meta-analysis establishing the role of MRD in all disease settings
Munshi NC, et al. Blood Adv. 2020;4(23):5988-5999.
61
Next phase in the search for a CURE Molecular and immune testing
Best Frontline Therapy
MRD Positive or Biochemical relapse
New Immune Therapy Cocktail
62
Opportunities for 2021-2023 New Trials To achieve MRD undetected at biochemical relapse Cutting edge disease monitoring Blood MRD sensitivity at 10-8 Mass spectrometry (high sensitivity) Serial immune testing Molecular characterization 63
Frontline Therapy A dilemma will continue: Triplets (VRd/ VTd/ KRd) Versus
Quadruplets + Dara/Isa Efficacy/ costs/ access/ toxicities will be debated
64
MAIA TRIAL: DRd vs Rd
T Facon et al. N Engl J Med 2019;380:2104-2115.
65
CASSIOPEIA TRIAL: Dara-VTd vs VTd Quadruplets as Initial Therapy
The Lancet 2019 394, 29-38DOI: (10.1016/S0140-6736(19)31240-1) Copyright © 2019 Elsevier Ltd Terms and Conditions
66
Upfront ASCT Will become priority if MRD positive
Will be assessed versus CAR T and other powerful immune therapies
67
Consolidation Consolidation will become more important
to achieve MRD undetected: Extra 2 cycles if needed (6 total) CAR T or other immune Rx New agents
68
Maintenance Maintenance will include:
Revlimid® Proteasome inhibitor Dara/ Isa With target for ~ 2 year duration
69
Early relapse (1-3 prior regimens) Use a triplet if feasible
Incorporate Dara/Isa early if not use previously Consider Kyprolis® if high risk New immune therapies may become top choices!
70
Later relapses (triple exposed) Multiple approved new agents such as:
Selinexor®, Belantamab, CAR T Multiple new agents in trials including: CelMods, bispecifics, new targeted agents
Studies with “relaxed” entry criteria required to assess real world data/ experience and evidence
and make agents available to poorer-risk patients 71
COVID will be with us! • Vaccination • Boosters • Masks • Other protections
72
Despite COVID… The future is bright with likely improved outcomes 73
Panel Discussion
74
Audience Q&A
• Open the Q&A window, allowing you to ask questions to the host and panelists. They can either reply to you via text in the Q&A window or answer your question live. • If the host answers live, you may see a notification in the Q&A window.
• If the host replies via the Q&A box – you will see a reply in the Q&A window.
IMF Patient and Family Webinar
76
IMF Patient and Family Webinar Agenda
*times listed in US Eastern Time Zone
11:30 – 12:00 PM
Immune Therapies: Treatments That Work With Our Own Immune Systems Dr. Adam Cohen
12:00 – 12:20 PM
How to Find What You Need to Know Robin Tuohy, IMF Vice President, Support Groups
12:20 – 12:50PM
When Myeloma Comes Back: Approaches to Relapse Dr. Rafat Abonour
12:50 – 1:00 PM
Summary Panel Discussion Webinar Survey & Closing Remarks 77
IMF Patient and Family Webinar
Adam Cohen, MD
Immune Therapies: Treatments That Work With Our Own Immune Systems
Abramson Cancer Center University of Pennsylvania Philadelphia, PA
78
Immunotherapies: CAR T cells and Bispecific Antibodies for Multiple Myeloma Adam D. Cohen, MD Abramson Cancer Center University of Pennsylvania Philadelphia, PA, USA IMF Patient and Family Webinar
October 30, 2021
Division of Hematology/Oncology
Disclosures ‣ Consulting/Advisory Boards: Celgene, BMS, Takeda, Janssen, Seattle Genetics, AstraZeneca, Genentech/Roche, Oncopeptides, GlaxoSmithKline ‣ Research support: Novartis, GlaxoSmithKline ‣ Intellectual property licensed by institution to: Novartis
80
Types of Immunotherapy Directly targeting myeloma cell markers
Boosting myelomafighting T cells
Monoclonal and Bispecific antibodies
CAR T cells
Rodriguez-Otero P et al. Haematologica. 2017;102:423.
IMiDs/CelMODs, checkpoint inhibitors
Vaccines
Overcoming immune suppression
Activating myelomaspecific immunity
81
T cells can recognize and kill cancer cells BUT…in patients • Cancer cells learn to evade the T cells • T cells lose their activity over time T cell HOW TO OVERCOME THIS? • Checkpoint blockade • Cellular therapy • Bispecific antibodies/T cell engagers (BiTEs)
Groscurth P, Filgueira L. Physiology. 1998;13:17-21.
From CAR T Working Group, ASTCT slide library
82
CAR (Chimeric Antigen Receptor)
Chimera
‣ Combines recognition domain of antibody with signaling domain of T cell ‣ Uses gene transfer (eg. lentiviral vector) to stably express CAR on T cells allows recognition of cancer cell
‣ Addition of co-stimulatory domains (CD28, 4-1BB) augments proliferation and survival of the T cells 83
Building CAR T cells T cell
CTL019 cell Lentiviral vector
1. Cytotoxicity 2. Cytokine production 3. Long-term persistence
“Living drug”
Native TCR Anti-CD19 CAR construct
CD19
Dead tumor cell
Tumor cell
Courtesy of D. Porter
84
Overview of CAR T cell therapy
Courtesy of D. Porter
85
CD19-specific CAR T cells for B-cell cancers ‣ Acute lymphoblastic leukemia (ALL) ‣ Chronic lymphocytic leukemia (CLL) ‣ B-cell Non-Hodgkin lymphomas ‣ Initial trials reported in 2010-2011 • • • •
Memorial Sloan Kettering Cancer Center/New York University of Pennsylvania/Philadelphia National Institutes of Health/Bethesda Fred Hutchinson Cancer Center/Seattle
4 FDA approved CD19 CAR T products: Kymriah Yescarta Tecartus Breyanzi
‣ Dramatic responses seen in highly-refractory patients • Can be durable, >10 years in some earliest-treated patients https://vimeo.com/54668275 www.emilywhiteheadfoundation.org 86
CAR T cell Toxicity 1. CRS (>80% of patients; <10% Gr3-4) 1. 2. 3.
4.
Occurs between 1 hour and 10 days after infusion Lasts between 3-5 days Symptoms: 1.
Fever, chills, HA, fatigue and malaise [flu-like]
2. 3.
Low blood pressure, fast heart rate Shortness of breath, need for oxygen,
Treatment: tocilizumab +/- dexamethasone
2. Neurotoxicity (<30%; <10% G3-4) 1. 2. 3.
Occurs between 2 days to ~30 days, some later Lasts between 3-14 days, few have longer symptoms Symptoms 1. 2. 3.
Confusion, delirium, aphasia Tremor, Parkinson-like symptoms, nerve palsies Brain swelling is rare
3. Cytopenias 1. 2. 3.
Low WBC Low platelets Anemia
4. Infection 1.
Viral, Bacterial, fungal and unusual infections
87
BCMA (B-cell Maturation Antigen): a new target for myeloma ‣ Expressed on normal plasma cells ‣ Highly expressed on myeloma cells ‣ Soluble BCMA in patient serum Promotes MM growth and survival Multiple approaches targeting BCMA • Antibody-drug conjugates • Bispecific Antibodies • CAR T cells
Frigyesi et al, Blood 2014; Tai et al, Blood 2014; Carpenter et al, Clin Can Res 2013; Tai et al, Blood 2016
88
BCMA CAR T cells highly active in relapsed/refractory myeloma
Studies started 2015 2016
# lines
% hi risk†
Dosing
ORR
ORR (optimal doses)
VGPR/CR (optimal doses)
Trial
n
CAR
Conditioning
NCI1
26*
Murine, CD3/CD28
Cy/Flu
7.5
42%
0.3 – 9 x 106/kg
58%
81% (13/16)
63% (10/16)
Penn2
25
Human, CD3/41BB
None or Cy
7
76%
0.5 – 5 x 108
48%
64% (7/11)
36% (4/11)
Bluebird bb21213
33
Murine, CD3/41BB
Cy/Flu
7.5
40%
0.5 – 8 x 108
85% (28/33)
90% (27/30)
80% (24/30)
Legend4
57
Llama, CD3/41BB
Cy
3
??
0.07-2.1 x106/kg
88%
88% (50/57)
74% (42/57)
*2 treated twice; counted separately for response. † FISH +t(4;14), t(14;16), del 17p Cy = cyclophosphamide, Flu = fludarabine
‣ BCMA CAR T cell products: • Idecabtagene vicleucel (ide-cel) Abecma (FDA approved) • Ciltacabtagene autoleucel (cilta-cel) (under FDA review)
1Brudno,
J Clin Oncol 2018; 2Cohen, J Clin Invest 2019; 3Raje, NEJM 2019; 4Zhou, J Hematol Oncol 2018
89
Ph 2 - KarMMa-1: Study Design
bb2121 CAR Design MND SP Anti-BCMA scFv Promot er
CD8 Linke r
Tumor binding domain
4-1BB
CD3z
Signaling Domains
90
Ide-cel Median 6 prior lines, 84% triple-refractory
52% got tocilizumab
CAR, chimeric antigen receptor; CR, complete response; CRR, complete response rate; CRS, cytokine release syndrome; CTCAE, common criteria for adverse events; MRD, minimal residual disease; NE, not evaluable; ORR, overall response rate; PR, partial response; sCR, stringent complete response; CAR
Munshi et al. ASCO 2020; abstract 8503 (oral presentation)
91
92
Ciltacabtagene autoleucel (cilta-cel; JNJ-68284528; LCAR-B38M)
‣ Madduri et al. ASH 2020; abstract 177 (oral presentation)
93
CARTITUDE-1 (cilta-cel) updated phase 1/2 data Median 6 prior lines, 88% triple-refractory
FDA review date 11/29/21 ‣ Usmani et al. ASCO 2021; #8005
94
Ongoing ide-cel and cilta-cel CAR T cell trials Ide-cel ‣ KarMMa-2 • Phase 2 study in r/r MM and high-risk MM (relapse early after induction)
‣ KarMMa-3 • Phase 3 randomized study of ide-cel vs SOC in r/r MM
‣ KarMMa-4 • Phase 1 study in newly diagnosed high-risk MM
Cilta-cel ‣ CARTITUDE-2 • Phase 2 study in multiple cohorts – – – – – –
A: 1-3 prior therapies B: Relapse early after induction +/- SCT C: After prior BCMA-targeted treatment D: <CR after SCT (combo with Revlimid maint) E: newly-diagnosed high risk F: standard risk after induction therapy
‣ CARTITUDE-4 • Phase 3 randomized study of cilta-cel vs SOC in r/r MM
95
Other CAR-based approaches in myeloma ‣ Autologous BCMA CAR T cells • • • • • • •
JCARH125 (orva-cel) FCARH125 P-BCMA-101 bb21217 CT053 FHVH-BCMA-T CT103A
‣ Bispecific CAR T cells
‣ “Off-the-shelf” allogeneic CAR products • • • • • • •
ALLO-715 ALLO-605 UCARTCS1 CTX120 CYAD-211 PBCAR269A P-BCMA-ALLO1
• FT576 (CAR-NK)
• GCF012 (CD19/BCMA-targeted) • BM38 (BCMA/CD38-targeted) • BCMA/GPRC5D (pre-clinical) Mailankody et al, ASH 2020, #129; Goodridge et al, ASH 2020, #1402
96
Types of Antibodies (Antibody-drug conjugate)
97
Bispecific Antibodies/T-cell Engagers for Myeloma
AMG420
Shah N et al, Leukemia 2020; www.clinicaltrials.gov (search 10/09/21)
98
Phase 1 study of Teclistamab (BCMA x CD3 bispecific Ab)
Krishnan et al, ASCO 2021, #8007
99
Phase 1 study of Elranatamab (BCMA x CD3 bispecific Ab)
Bahlis et al, ASCO 2021, #8006
100
Beyond BCMA?
Chari et al. ASH 2020; #290; Cohen et al. ASH 2020; #292
101
Redirecting T cells: CARs vs. Bispecific Antibodies CAR T cells
‣ Advantages • • •
One-time treatment Toxicities front-loaded Durable remissions seen
Bispecific Abs
‣ Advantages • • • •
Off-the-shelf Lower frequency/severity of CRS, ICANS No lymphodepletion needed Durable remissions seen
‣ Disadvantages •
Manufacturing headaches –
•
Need for lymphodepletion –
•
Prolonged cytopenias, risk of MDS
Higher frequency/severity of CRS, ICANS –
•
slot allocation, need for apheresis, manufacturing failures, disease progression
Need for hospitalization
‣ Disadvantages • •
Response rates a bit lower Repeated (?indefinite) administration –
• •
Ongoing risk of toxicity
Hospitalization for step-up dosing Shorter follow-up in trials to date
Not “curative” in triple class-refractory setting
102
Sequencing Bispecific Abs with CAR T cells Apheresis
BCMA BsAb
BCMA CAR
Apheresis
Non-BCMA BsAb
BCMA CAR
BCMA BsAb
Apheresis
BCMA BsAb
BCMA CAR
Non-BCMA BsAb
Apheresis
Non-BCMA BsAb
BCMA CAR
BCMA CAR
BCMA BsAb
BCMA CAR
Non-BCMA BsAb
103
Summary/Take-home points ‣ BCMA CAR T cells induce deep responses in relapsed/refractory MM patients • Registration studies completed for ide-cel (Abecma) and cilta-cel – Phase 3 studies vs standard regimens and Phase 2 studies in less heavily-treated pts ongoing – Durability of responses (median PFS 12-24 months) good but needs improvement – CRS and neurotoxicity remain issues but high grade uncommon (≤10%)
• Ongoing phase 1 studies for next-generation autologous CAR T products • Proof-of-principle safety/activity with off-the-shelf allogeneic BCMA CAR product (ALLO-715) – CAR-NK cell products to start trials in 2021/2022
‣ BCMA and non-BCMA-targeted bispecific antibodies/T cell-engagers (BsAb/BiTEs) induce deep responses in relapsed/refractory MM patients • Response durability promising but follow-up shorter • Optimal dose/schedule to be determined
‣ Challenges: choosing between products, optimal sequencing, combining with standard myeloma drugs
104
IMF Patient and Family Webinar
How to Find What You Need to Know Robin Tuohy International Myeloma Foundation Vice President, Support Groups 105
IMF Patient and Family Webinar
When Myeloma Comes Back: Approaches to Relapse Paul Richardson, MD Dana-Farber Cancer Institute Boston, MA
106
The Evolving Role of Novel Therapy in the Treatment Paradigm of Relapsed and Refractory Multiple Myeloma Paul G. Richardson, MD RJ Corman Professor of Medicine Harvard Medical School Clinical Program Leader, Director of Clinical Research Jerome Lipper Multiple Myeloma Center Dana-Farber Cancer Institute Boston, Massachusetts
Boston MA 2021
Disclosures • Advisory Committees
• Celgene/BMS, Takeda, Janssen, Oncopeptides, Sanofi, Secura-Bio, Karyopharm, GSK, Regeneron • Research Funding
− Celgene/BMS, Takeda, Oncopeptides
Multiple Novel Agents Are Now Available to Treat Newly Diagnosed and Relapsed/Refractory Myeloma 2021 How do we sequence and strategize therapies to ensure the best outcomes for our patients?
15 Approved Novel Agents in MM…..and more coming!
Doxil
Lenalidomide
Pomalidomide
Bortezomib
Ixazomib
Thalidomide
Carfilzomib
Daratumumab
Elotuzumab
1st line
2nd line st
1 line
? Panobinostat
Selinexor
Melflufen
Isatuximab
3rd line Andnd 2 line beyond
Belamaf
Adapted from Laubach JP et al. Leukemia. 2016. Moreau P et al. Lancet Oncol. 2021. Idecel
Treating Multiple Myeloma Is a Marathon, Not a Sprint!
Strategic vs. tactical considerations ~ Tolerability AND efficacy key with combination regimens
Adapted from Borrello I. Leuk Res. 2012;36:S3 Richardson PG, San Miguel J et al. Blood Cancer Journal 2018 “Translating Clinical Trial Results to Real World Practice”.
Key Targets in MM 2021 Genomic abnormalities: • Target and overcome mutations • Critical Role of Combination and Continuous Therapy • Evolving Position and Timing of ASCT Excess Protein Production: • Target Protein Degradation Immune Suppression: • Restore anti-MM immunity Richardson PG et al, MMRF 2021
112
Definition of RRMM • IMWG criteria for PD1 • ≥25% increase from nadir in: • Serum/urine M-protein (absolute increase ≥0.5 g/dLa and ≥200 mg per 24 h, respectively) or • Difference between involved and uninvolved FLC levelsb (absolute increase >100 mg/L) or • BM plasma cellsc (absolute increase ≥10%) or • New lesions (≥50% increase in SPD of >1 lesion or longest diameter of previous lesion >1 cm in short axis) or • Circulating plasma cells (≥50% increase [minimum 200 cells/μL] if only measure of disease)
• Criteria for RRMM2-5 • Meets IMWG criteria for PD1 • RRMM: progression on tx in patients who obtain at least minor response or progression within 60 days on most recent tx • Primary refractory MM: not achieving at least minor response on a given tx • Relapsed MM: meets IMWG criteria for PD but not RRMM or primary refractory MM
BM, bone marrow; FLC, free light chain; IMWG, International Myeloma Working Group; MM, multiple myeloma; PD, progressive disease; SPD, sum of the products of the maximal perpendicular diameters of measured lesions. If lowest M component ≥5 g/dL, increase must be ≥1 g/dL; b In patients without measurable serum/urine M-protein; c In patients without measurable serum/urine Mprotein or involved FLC. a
1. Kumar. Lancet Oncol. 2016;17:e328. 2. Nooka. Blood. 2015;125:3085. 3. Rajkumar. Blood. 2011;117:4691. 4. Richardson PG NEJM 2003. Richardson PG, et al. Cancer. 2006;106:1316-1319.
113
Median Response Duration (mos)
Confronting Disease Relapse in Myeloma Response Duration With Increasing Treatment1 12 10 8 6 4
2 0 First Second Third
Fourth
Fifth
Treatment Regimen EFS, event-free survival.
1. Kumar. Mayo Clin Proc. 2004;79:867. 2. Bahlis. Blood 2017
Sixth
Novel Factors Impacting Treatment Choice in RRMM: Refractoriness to Drug Classes Real-World Progression-Free Survival (months)
• •
•
Outcomes of patients have improved with the advent of new therapies However, as the disease relapses, it becomes increasingly refractory to available treatments, resulting in shorter remissions Quality and duration of response to previous therapies are therefore important prognostic factors when treating patients with RRMM
Figures reproduced from Bruno A. S., et al. Expert Review of Hematology 2020.1 Newer treatments (approval date during/after 2012) were pomalidomide, carfilzomib, elotuzumab, panobinostat, daratumumab, ixazomib, and bortezomib lenalidomide combination rwOS, real-world overall survival; rwPFS, real-world progression-free survival. 1. Bruno A.S. et al., Expert Rev Hematol. 2020 Sep;13(9):1017-1025.
Real-World Overall Survival (months)
115
Effect of Drug Class Refractoriness Double refractory EFS and OS Outcomes1 100
Patients (%)
80
Events, n/N
Median, mos (range)
OS
170/286
9 (7-11)
EFS
217/286
5 (4-6)
60 40 20 0 0
12
24
36
48
Months
OS, overall survival. EFS, event-free survival 1. Kumar. Leukemia. 2012;26:149. 2. Gandhi. Leukemia. 2019;33:2266.
60
Double, triple, quad and Penta- refractory OS2
116
When Should We Start Treatment for Relapse? • Patients with clinical progression/CRAB clearly need treatment • Those with biochemical progression only may not need immediate treatment, or can consider a rational addition to current therapy • Standard-risk disease with slow trend upward CRAB, hypercalcemia, renal failure, anemia, bone disease.
Presentation with renal or neurological complications
High-risk disease with any progression
Rapid doubling of M spike
Indications for treatment
Adapted from Laubach et al , Leukemia 2016 Moreau et al, Lancet Oncol 2021
IMW 2021 How Are New Drug Classes Affecting the Treatment Landscape in RRMM? A Complex Question For a Complex Series of Issues
Triple- or quadruplea-class or penta-drugb refractory
Renal dysfunction Bone marrow reserve Immune deficiency (including T-cell dysfunction)
Elderly and frail
Neuropathy
•
aDefined
•
CD, cluster of differentiation; IMiD, immunomodulatory drug; mAb, monoclonal antibody; PI, proteasome inhibitor.
as refractory to ≥1 IMiD, 1 PI, 1 anti-CD38 mAb and 1 glucocorticoid; bDefined as refractory to ≥2 PIs, ≥2 IMiDs and 1 anti-CD38 antibody.
Recommended Regimens in RRMM (adapted from NCCN guidelines 2020-2021) Treatment at first relapse
Not refractory to LENa
Treatment after multiple relapses
Refractory to LEN
Considerb
DRd1 or KRd2, PVd 9
Frail: IRd,3 ERd4
DVd,5 DPd,6 DKd,7 KPD8 PVd9, EloPd10 Panobinostatbased rx16
Any first relapse options not yet used (2 new drugs; triplet preferred) Additional approved options: Isatuximab-combinations selinexor,15 VTd-PACE, bendamustine-based rx, Panobinostat-based rx16
Frail: Pd,11 IPd,12 VCd,13 PCd14
Investigational options: BCMA-targeting therapy on clinical trial; melflufen17,18, venetoclax
ASCT, autologous stem cell transplant; BCMA, B-cell maturation antigen; BTZ, bortezomib; CFZ, carfilzomib; DKd, dara, CFZ, and DEX; dara, daratumumab; DPd, dara, POM, and DEX; DRd, dara, LEN, and DEX; DVd, dara, BTZ, and DEX; EloPd, elotuzumab, POM, and DEX; ERd, elotuzumab, LEN, and DEX; IPd, ixazomib, POM, and DEX; IRd, ixazomib, LEN, and DEX; LEN, lenalidomide; PCd, POM, cyclophosphamide, DEX; Pd, Pm, DEX; POM, pomalidomide; VCd, BTZ, cyclophosphamide, DEX; VPd, BTZ, POM, DEX; VRd, BTZ, LEN, DEX; KPd, CFZ, POM, DEX; KRd, CFZ, LEN, DEX; KCd, CFZ, cyclophosphamide, and DEX; VDT-PACE, BTZ, DEX, thalidomide, cisplatin, doxorubicin, cyclophosphamide, and etoposide. a
Relapse occurring while off all rx, on low doses of single-agent LEN, or on BTZ maintenance; b For those intolerant of LEN, on higher doses of single-agent LEN, with aggressive relapse, or with highrisk disease; c Consider salvage ASCT in eligible patients if first PFS >2 y off maintenance or >4 y with maintenance, or for patients with pancytopenias and heavy BM involvement.
1. Dimopoulos. Haematologica. 2018;103:2088. 2. Stewart. ASH 2017. Abstr 743. 3. Moreau. NEJM. 2016;374:1621. 4. Lonial. ASCO 2018. Abstr 8040. 5. Lentzsch. ASCO 2017. Abstr 8036. 6. Chari. Blood. 2017;130:974. 7. Chari. ASCO 2018. Abstr 8002. 8. Bringhen. Leukemia. 2018;32:1803. 9. Richardson PG et al . Lancet Oncol 2019. 10. Dimopoulos. EHA 2018. Abstr LBA2606. 11. San Miguel. Lancet Oncol. 2013;14:1055. 12. Krishnan. Leukemia. 2018;32:1567-1574. 13. Rajkumar. Am J Hematol. 2014;89:999. 14. Baz. Blood. 2016;127:2561. 15. Vogl. J Clin Oncol. 2018;36:859. 16. Laubach JP et al , Lancet Oncol 2020. 17. Richardson PG et al. ASH 2018. Abstr 600. 18. Richardson PG et al, J Clin Oncol 2021.
IMW 2021 - Improving Outcomes for Patients with RRMM: Patient Perspective •
There are multiple factors of importance to MM patients regarding their treatment that can have an impact on the effectiveness of that treatment in the real-world setting1
Factors of importance to patients in the real-world setting1
Burden
Considerations for optimal treatment
Symptom burden
Symptoms related to the CRAB criteria can be debilitating and may require supportive therapy
Rapid symptom control
Side effects
Side effects can affect treatment adherence and lead to treatment discontinuation
Treatment options with minimal toxicity
Disturbance of daily life
Impairment of activities of daily living due to MM treatment or other comorbidities can be associated with poorer prognosis
The ability to continue with one’s daily routine and physical activities while receiving treatment
Financial toxicity
Direct out-of-pocket costs arising from treatment and its side effects, travel costs
Patient’s ability to cope with the financial toxicity associated with receiving their regimen on a longterm basis
Treatment convenience /route of administration
Cost and logistics associated with frequent hospital/clinic visits
Preference for oral treatment
Traditional Factors Impacting Treatment Choice in RRMM: Frailty Status TTNT in intermediate to frail patients
• Advanced age, functional decline, and comorbid conditions represent components of frailty that are predictive of mortality and toxicity risk in MM
• In the real-world, frailty can have a significant impact on the effectiveness of different treatment options
Traditional Factors Impacting Treatment Choice in RRMM: Extramedullary Disease •
EMD is highly prevalent in RRMM patients and is associated with poor prognosis, making the management of this patient group particularly challenging •
•
Prognosis is usually poorer for patients with hematogenous-spread soft-tissue plasmacytomas than for patients with bone-related plasmacytomas
No standard therapy has been established for this population with high unmet medical need •
Treatments with Selinexor, Isatuximab, Melflufen and CAR T-cell therapy as well as others have shown promising efficacy in patients with RRMM and EMD OS in EMD vs Non-EMD Groups: Melflufen
OS in EMD vs Non-EMD Groups: CAR T-cell Therapy
122
Agents in Relapsed MM Lenalidomide-Based Studies POLLUX DRd vs Rd1
ASPIRE KRd vs Rd2
ELOQUENT-2 ERd vs Rd3,4
TOURMALINE-MM1 IRd vs Rd5
PFS HR (95% CI)
0.37 (0.27-0.52)
0.69 (0.57-0.83)
0.73 (0.60-0.89)
0.74 (0.59-0.94)
ORR, %
93
87
79
78
≥VGPR, %
76
70
34
48
≥CR, %
43
32
5
14
DoR, mos
NE
28.6
20.7
20.5
0.64 (0.40-1.01)
0.79 (0.63-0.99)
0.77 (0.61-0.97)
NE
Outcomes
OS HR (95% CI)
CI, confidence interval; CR, complete response; DoR, duration of response; HR, hazard ratio; NE, not estimable; PFS, progression-free survival; Rd, LEN and DEX; VGPR, very good partial response. 1. Dimopoulos. EHA 2016. Abstr LB238. 2. Stewart. NEJM. 2015;372:142. 3. Lonial. NEJM. 2015;373:621. 4. Dimopoulos. ASH 2015. Abstr 28. 5. Moreau. NEJM. 2016;374:1621.
123
Agents in Relapsed MM PI-Based Studies CASTOR DVd vs Vd1
ENDEAVOR Kd vs Vd2
Panobinostat PVd vs Vd3,4
OPTIMISMM PomVd vs Vd5
PFS HR (95% CI)
0.39 (0.28-0.53)
0.53 (0.44-0.65)
0.63 (0.52-0.76)
0.61 (0.49-0.77)
ORR, %
83
77
61
82
Median PFS, mos
NR
18.7
12.0
11.2
≥VGPR, %
59
54
28
53
≥CR, %
19
13
11
12.5
DoR, mos
NE
21.3
13.1
13.7
0.77 (0.47-1.26)
0.79 (0.58-1.08)
0.94 (0.78-1.14)
0.98 (0.73-1.32)
Outcomes
OS HR (95% CI)
Kd, CFZ and DEX; NR, not reached; PVd, POM, BTZ, and DEX. 1. Palumbo. N Engl J Med. 2016;375:754. 2. Dimopoulos. Lancet Oncol. 2016;17:27. 3. San-Miguel. Lancet Oncol. 2014;15:1195. 4. San-Miguel. ASH 2015. Abstr 3026. 5. Richardson PG, et al. Lancet Oncol. 2019;20:781-794.
OPTIMISMM: PVd versus Vd in RR MM Study Design1,2 Phase 3 trial comparing the efficacy and safety of PVd vs Vd in LEN-exposed RRMM patients. Funded by Celgene Corporation
21-day cycles
LT follow-up
PVd (n = 281) POM BORT
RRMM • 1-3 prior regimens, ≥ 2 cycles of LEN • ECOG PS ≤ 2 • Prior BORT allowed (PD with 1.3 mg/m2 twice weekly dose excluded)a N = 559
LoDEX
R 1:1
4 mg Days 1-14 1.3 mg/m2 SC Cycles 1-8: Days 1, 4, 8, 11 Cycles 9+: Days 1 and 8 20 mg (≤ 75 years) or 10 mg (> 75 years) day of and day after BORT
Vd (n = 278) BORT
LoDEX
Tx discontinued due to PD
Follow-up visit 28 days after Tx discontinuation PD or unacceptable toxicity
1.3 mg/m2 SC Cycles 1-8: Days 1, 4, 8, 11 Cycles 9+: Days 1 and 8 20 mg (≤ 75 years) or 10 mg (> 75 years) day of and day after BORT
PD, subsequent antimyeloma Tx, and survival Tx discontinued prior to PD
Enter PFS follow-up periodb
NCT01734928
Stratification • Age (≤ 75 years vs > 75 years) • Prior regimens (1 vs > 1) • β2-microglobulin at screening (< 3.5 mg/L vs ≥ 3.5 to ≤ 5.5 mg/L vs > 5.5 mg/L)
1°
PFS (time from randomization to disease progression or death)
2°
OS, ORR by IMWG criteria, DOR, safety
Data cutoff: • October 26, 2017
Study Endpoints
TTR, PFS2, efficacy analysis in
Key subgroups, including refractoriness to exploratory LEN and number of prior lines of therapyc, and HRQoL
a Patients
with PD during therapy or within 60 days of the last dose of a BORT-containing therapy under the approved dosing schedule of 1.3 mg/m2 twice weekly were excluded. b Efficacy evaluated every 21 days (± 3 days) until PD. c These subgroups were predefined in the statistical analysis plan.
1. Dimopoulos M, et al. Poster presented at ASH 2018 [abstract 3278]. 2. Weisel K, et al. Poster presented at ASH 2018 [abstract 1960].
OPTIMISMM: PVd VERSUS Vd IN RRMM PFS ITT AND 1 PRIOR LINE
Response
OPTIMISMM: PVd Versus Vd In RRMM Overall Summary •
•
Study Design • OPTIMISMM is an open-label, randomized, Phase 3 study comparing the efficacy and safety of PVd vs Vd in LEN-pretreated patients with RRMM Results • Primary Efficacy Data • In the ITT population, the median PFS with PVd vs Vd was 11.20 months vs 7.10 months (HR = 0.61 [95% CI, 0.49-0.77]; P < .0001) • In LEN-refractory patients, the median PFS was 9.53 vs 5.59 months (HR = 0.65 [95% CI, 0.50-0.84]; P < .0008) • In patients with 1 prior line of therapy, the median PFS was 20.73 vs 11.63 months (HR = 0.54 [95% CI, 0.36-0.82]; P < .0027) • ORR was 82.2% vs 50.0% (P < .0001) in the ITT population • Safety Data • Most common Grade ≥ 3 hematologic AEs with PVd included neutropenia (42% vs 9% with Vd) and thrombocytopenia (27% vs 29% with Vd) • Most common Grade ≥ 3 nonhematologic AE with PVd was infection (31% vs 18% in the Vd arm); all other Grade ≥ 3 nonhematologic AEs occurred in < 10% of patients • Incidence of SPMs was 3% (2.7 per 100 person-years) with PVd vs 1% (1.2 per 100 person-years) with Vd • A total of 86 patients died in each arm; mostly due to myeloma progression and infections • Discontinuation of the lead drug were reported in 11% in the PVd and 19% of patients in the Vd arm, respectively Richardson PG, et al. Lancet Oncol. 2019; published online May 13. DOI:https://doi.org/10.1016/S1470-2045(19)30152-4.
Panobinostat: HDAC Inhibition for the treatment of RR MM • Panobinostat’s unique multi-modal MOA alters gene expression patterns both directly and via histone modification to overcome resistance to other therapies1-3 Panobinostat
Activation of tumor suppressor genes
Histone-Driven (epigenetic)
Direct (Non-histone) HDAC
HDAC1,2,3
HDAC1,2
Histones
p53
HDAC6
Tubulin
HSP90
Aggresome
Image adapted from Laubach JP, et al. Clin Cancer Res. 2015;21(21):4767-4773. 1. Richardson PG, et al. Blood. 2013;122:2331-2337; 2. Imai Y, et al. Cancers (Basel). 2019;11(4):475; 3. Yeon M, et al. Front Cell Dev Biol. 2020;8:486.
Accumulation of misfolded proteins (critical to MM)
Cell Death
Duration of Response in RR MM: Pano Vd PANORAMA-3 Responders without event, %
100 80
PAN 10 mg TIW PAN 20 mg BIW PAN 20 mg TIW
Median DoR Event months Patients Events Censored rate (95% CI) 44 22 22 50% 10.84 (6.24, 14.49) 56 26 30 46% 11.76 (8.77, 21.26) 51 19 32 37% 22.08 (13.86, NE)
60 40 PAN 10 mg TIW PAN 20 mg BIW PAN 20 mg TIW
20 0 0
At risk, n PAN 10 mg TIW PAN 20 mg BIW PAN 20 mg TIW
44 56 51
2
4
6
8
39 49 49
33 36 37
23 32 34
17 25 27
10
14 20 23
12
8 11 21
14
6 10 17
16 18 20 Time, months 4 6 13
3 3 12
3 3 12
22
3 2 11
24
26
28
30
32
34
36
1 2 8
0 2 6
1 6
1 3
0 1
1
0
Laubach JP et al, Lancet Oncology 2020
Monoclonal Antibody-Based Approaches
With a focus on targeting CD38 and SLAMF7
131
CANDOR: Response Rates
ORR
KdD (n=312)
Kd (n=154)
84.3a
74.7a
≥VGPR
69.2
48.7
≥CR
28.5
10.4
MRD negative at 12 mos (10–5 threshold)
17.6
3.9
MRD negative CR at 12 mos (10–5 threshold)
12.5b
Best MRD negative CR (10–5 threshold)
1.3b
Median PFS: NE vs 15.8 mos 1.0 + ++ + +
Proportion Surviving Without Progression
Response, %
+ ++
0.8
0.6
0.4
+++ ++ ++++++++++ +++ + ++ + +++++ ++ + KdD ++ ++ ++++++++++++++++++ ++ + + + + + + + + + + + +++++++++++++ ++ + ++++ ++ + +++ + + +++++++ +++++ ++ +++++++++ +
Kd 0.2
0.0
13.8
3.2
0
3
6
9
12
15
18
21
24
Months After Randomization At risk, n KdD
302
279
236
211
189
165
57
14
0
Kd
154
122
100
85
70
55
13
2
0
KdD, CFZ, DEX, and Dara. a
P=0.0040; b P<0.0001. Usmani. ASH 2019. Abstr LBA6. Lancet 2020
132
CANDOR: PFS • Prolonged PFS with KdD vs Kd • Median, NR vs 15.8 mo • HR, 0.63 • 95% CI, 0.46-0.85
Subgroup
Subgroup
HR KdD vs Kd (95% CI)
ISS stage
• 1 or 2 •3
0.61 (0.43-0.85) 0.71 (0.37-1.36)
Cytogenic risk group
Baseline age, y
• ≤64 • 65-74 • ≥75
0.57 (0.38-0.86) 0.72 (0.43-1.20) 0.97 (0.39-2.43)
• High • Standard • Unknown
0.58 (0.30-1.12) 0.55 (0.31-0.97) 0.72 (0.47-1.11)
Number of prior tx
•1 • ≥2
0.70 (0.42-1.17) 0.63 (0.44-0.92)
Region
• North America • Europe • Asia Pacific
0.04 (0.01-0.34) 0.86 (0.60-1.23) 0.49 (0.25-0.93)
Prior LEN exposure
• No • Yes
0.87 (0.56-1.35) 0.52 (0.34-0.80)
Baseline ECOG PS
• 0 or 1 •2
Refractory to LEN
0.69 (0.51-0.94) 0.31 (0.08-1.19)
• No • Yes
0.85 (0.57-1.27) 0.45 (0.28-0.74)
Baseline CrCl, mL/min
• ≥15 to <50 • ≥50 to <80 • ≥80
0.46 (0.21-1.02) 0.78 (0.45-1.33) 0.67 (0.44-1.02)
Prior PI exposure
• No • Yes
0.93 (0.29-3.02) 0.64 (0.47-0.88)
Refractory to BTZ
• No • Yes
0.59 (0.40-0.85) 0.83 (0.49-1.41)
• P=0.0014)
Usmani. ASH 2019. Abstr LBA6. Lancet 2020
HR KdD vs Kd (95% CI)
EQUULEUS: Dara/POM/DEX in RRMM •
Single-arm phase 1b study (N=103; median of 4 prior lines [range: 1-13]; 71% refractory to PIs and IMiDs; 25% with high-risk cytogenetics
ORR (%)
60 50 40
ORR, 60% 17% CR or better
8% 9% 25%
30 20 10 0
42% VGPR or better
PR VGPR CR sCR
18%
Dara + POM/DEX (N=103)
• Of 17 patients in ≥CR, 35%, 29%, and 6% were MRD-negative at thresholds of 10–4, 10–5, and 10–6, respectively • Median OS, 17.5 mos (95% CI, 13.3-NE)
Chari A. et al. Blood. 2017;130:974.
PFS
100 80
PFS (%)
70
60 40 Median: 8.8 mos (95% CI: 4.6-15.4)
20 0
0
At risk, n 103
3
6
71
53
12 9 Months 42 28
15
18
21
12
1
0
• Median PFS with high-risk cytogenetics (n=22), 3.9 mos (95% CI, 2.3-NE)
134
MM-014: Phase I/II trial with Dara-Pd in Relapsed and/or Refractory MM patients based on len-refractory patients • 112 pts after 1 or 2 PL: all pts received prior Len, and 78% received prior Bz; 75% were refractory to most recent prior Len-containing regimen
APOLLO Phase 3 trial, Dara-Pd vs Pd Confirmed these Results….. Siegel et al. Leukemia 2020
APOLLO: Phase 3 Trial of SC Daratumumab, Pomalidomide and Dexamethasone (D-Pd) Versus Pd in RRMM 12-month PFS rateb
% surviving without progression
100
80
(PFS with median FU 16.9 mo)
60
52%
40
35%
D-Pd median: 12.4 months
20 HR, 0.63; 95% CI, 0.47-0.85; P = 0.0018
Pd median: 6.9 months
0 0 No. at risk Pd D-Pd
153 151
2
4
6
121 93 79 135 111 100
8
10
12
14
61 87
52 80
46 74
36 66
16 18 Months 27 48
17 30
20
22
24
26
28
30
32
34
36
12 20
5 12
5 8
1 5
0 3
0 2
0 2
0 2
0 1
Median PFS among patients refractory to lenalidomide was 9.9 months for D-Pd and 6.5 months for Pd
Addition of DARA SC to Pd improved PFS, with a 37% reduction in the risk of progression or death
Dimopoulos et al, ASH 2020
Phase III ICARIA-MM: Isatuximab + POM/DEX vs POM/DEX in RRMM Isa-Pd group • • • •
3 prior lines of tx 94% LEN refractory (60% in last line) 77% PI refractory 72% double refractory
Response
Pd
Isa-Pd
ORR, %
35
60
sCR
<1
0
CR
1
5
VGPR
7
27
PR
27
29
11.1
13.3
Median DoR, mos
Median follow-up
11.6 mos
Median PFS, mos
100
Isa-Pd Pd
80
PFS (%)
•
11.53 6.47
60 40 20
HR, 0.596 (95% CI, 0.436-0.814; P=0.001) 0
0 1 2
At risk, n Isa-Pd 154 Pd 153
129 105
3 4
5 6 7 8 9 10 11 12 13 14 15 16 Months 106 89 81 352 14 1 80 63 51 17 33 5 0
• PFS HR (95% CI) • LEN refractory, 0.59 (0.43-0.82) • LEN refractory in last line, 0.50 (0.34-0.76)
ISA, isatuximab.
Attal M, Richardson PG et al. Lancet. 2019;394:2096.
• LEN/PI refractory: 0.58 (0.40-0.84)
Isatuximab Plus Carfilzomib and Dexamethasone vs Carfilzomib and Dexamethasone in RRMM (IKEMA)
Interim PFS analysis of Isa-Kd compared with Kd showed hazard ratio of 0.531 At the time of analysis, overall survival data were immature Moreau et al, ASH 2020
138
IKEMA: Response 100 90 80 70 60 50 40 30 20 10 0
P=0.0004
Isa-Kd
P=0.19 86.6
MRD rate (NGS,a 10-5) 50
82.9 72.6
Isa-Kd
Kd
P=0.0011
41.4 40
56.1 39.7 27.6
Patients (%)
Incidence (%)
Best Overall Response (N=302)
29.6
30
22.9 20 13.0 10
ORR
VGPR or better
CR or better
53/179
16/123 MRD neg ITT
• Deeper responses with Isa-Kd, consistent with striking PFS improvement • MRD negativity rate with Isa-Kd, ~30% in ITT population
Moreau. EHA 2020. Abstr LB2603.
53/123
16/71
0 MRD neg in VGPR…
Kd
ELOQUENT-2: Efficacy Outcome
Elotuzumab + LEN/DEX (n=321)
LEN/DEX (n=325)
19.4
14.9
1 y, %
68
57
2 y, %
41
27
3 y, %
26
18
Median time to next tx, mos
33
21
ORR, %
79
66
43.7
39.6
HR (95% CI)
PFS Median, mos
Interim OS, mos
0.73 (0.60-0.89) P=0.0014
0.62 (0.50-0.77) 0.77 (0.61-0.97) P=0.0257
• PFS benefit seen with elotuzumab in all predefined subgroups
Dimopoulos M, Richardson PG et al. ASH 2015. Abstr 28.
Phase 2 ELOQUENT-3 : Elotuzumab/POM/DEX vs POM/DEX (N=117) PFS
80
60
Elotuzumab 40
Controls
20
OS
100
Surviving Patients (%)
Patients Surviving Without Progression (%)
100
Elotuzumab
80
60
Controls 40
20
HR, 0.54; 95% CI, 0.34-0.86; P=0.008 0
HR, 0.62; 95% CI, 0.30-1.28 0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22
Months Since Randomization
0
1
2
3
4
5
6
7
8
9 10 11 12 13 14 15 16 17 18 19 20 21 22
Months Since Randomization
Dimopoulos et al . NEJM. 2018;379:1811.
Selected Novel Targeted Agents for RR MM
IMW 2021: How Are New Drug Classes Affecting the Treatment Landscape in RRMM? A Complex Question For a Complex Series of Issues SINEs1
Peptide–drug conjugates3 Antibody–drug conjugates4
Peptide–drug conjugates9
CAR T-cell therapy2
Triple- or quadruplea-class or penta-drugb refractory
Renal dysfunction Bone marrow reserve
SINEs10
Antibody–drug conjugates11
BiTEs5
Bispecific antibodies6
Antibody–drug conjugates8
CELMoDs7
Elderly and frail
Immune deficiency (including T-cell dysfunction)
Neuropathy
Antibody–drug conjugates8
BiTEs12
•
aDefined
•
BiTE, bispecific T-cell engager; CAR, chimeric antigen receptor; CELMoD, cereblon E3 ligase modulator; SINE, selective inhibitor of nuclear export.
•
1. Bassali J, et al. Clinicoecon Outcomes Res. 2020;12:317-325; 2. Munshi NC, et al. N Engl J Med. 2021;384:705-716; 3. Richardson PG, et al. J Clin Oncol. 2021;39:757-767; 4. Becnel MR, Lee HC. Ther Adv Hematol. 2020;11:2040620720979813; 5. Harrison SJ, et al. Abstract 181. ASH Annual Meeting 2020; 6. Usmani SZ, et al. Lancet. 2021;398:665-674; 7. Van De Donk NWCJ, et al. Abstract 724. ASH 2020 Annual Meeting 2020; 8. Offidani M, et al. Drug Des Devel Ther. 2021;15:2401-2415; 9. Pour L, et al. Abstract 1028. EHA Annual Meeting 2021; 10. Grosicki S, et al. Lancet. 2020;396:1563-1573; 11. Lee HC, et al. J Clin Oncol. 2020;38(15_suppl):abstr 8519; 13; 12. Tian Z, et al. J Hematol Oncol. 2021;14:75.
as refractory to ≥1 IMiD, 1 PI, 1 anti-CD38 mAb and 1 glucocorticoid; bDefined as refractory to ≥2 PIs, ≥2 IMiDs and 1 anti-CD38 antibody.
Selective Inhibitors of Nuclear Export (SINEs)
SINEs mechanism of action1
SINEs XPO1 Myeloma cell
Tumour suppressor proteins
Selinexor Phase 3, open-label, global, randomised BOSTON Study (n=402)2 Eligibility criteria: • Aged ≥18 years • ECOG performance status of 0–2 • Previously received 1–3 lines of therapy
Proportion of patients (%)
Inhibition of XPO1 prevents transport of tumour suppressor proteins out of the cell nucleus, leading to their accumulation and activation, and therefore selectively inducing apoptosis of malignant cells1
Best overall response to SVd vs Vd in BOSTON2 100 sCR CR
80
60 40 20
n=19 n=14 n=54
n=62
VGPR
n=13 n=9 n=45
n=62
0
SVd (n=195)
Vd (n=207)
PR
SVd (n=195) or Vd (n=207)
1:1
SVd (n=195)
Vd (n=207)
HR (95% CI)
mPFS, months
13.93
9.46
0.70 (0.53–0.93) p=0.0075
mDOR, months
20.3
12.9
0.81 (0.56–1.17) p=0.1364
Most common Grade 3–4 AEs2 SVd (n=195)
Vd (n=204)
Thrombocytopaenia (39%)
Thrombocytopaenia (17%)
Anaemia (16%)
Pneumonia (11%)
Fatigue (13%)
Anaemia (10%)
Pneumonia (11%)
Fatigue (1%)
AE, adverse event; CI, confidence interval; CR, complete response; ECOG, Eastern Cooperative Oncology Group; HR, hazard ratio; mDOR, median duration of response; mPFS, median progression-free survival; PR, partial response; sCR, stringent complete response; SVd, selinexor, bortezomib, dexamethasone; Vd, bortezomib, dexamethasone; VGPR, very good partial response; XPO1, exportin 1. 1. Cheng Y, et al. Mol Cancer Ther. 2014;13:675-686; 2. Grosicki S, et al. Lancet. 2020;396:1563-1573.
Selective Inhibitors of Nuclear Export (SINEs) Selinexor Phase 3, open-label, global, randomised BOSTON Study of SVd vs Vd (n=402)1 A prespecified subgroup analysis for PFS revealed that the risk of progression or death was significantly lower in the SVd group than in the Vd group across a number of subgroups defined according to baseline characteristics: • • • •
Aged ≥65 years (HR 0.55 [95% CI 0.37–0.83]) High-risk cytogenetic abnormalities (HR 0.67 [95% CI 0.45–0.98]) One previous line of therapy (HR 0.63 [95% CI 0.41–0.95]) Previous treatment with lenalidomide (HR 0.63 [95% CI 0.41–0.97])
ORR was also significantly higher in the SVd group than in the Vd group for those patients: Subgroup
Proportion of patients (%)
Phase 2b, multicentre, open-label STORM Study of Sd (mITT n=122)2
100
Partial response or better to Sd in STORM2
80
sCR VGPR
60
PR
40 20
n=2 n=6
SVd
Vd
OR
p-value
Aged ≥65 years
76.1% [67.0–83.8]
64.4% (55.6–72.5)
1.77 (1.1–2.5)
0.0243
High-risk cytogenetic abnormalities
77.3% (67.7–85.2)
55.8% (45.2–66.0)
2.70 (1.4–5.0)
0.0008
Creatinine clearance 30–60 mL/min
79.2% (65.9–89.2)
56.7% (43.2–69.4)
2.92 (1.3–6.7)
0.0055
One previous line of therapy
80.8% (71.7–88.0)
65.7% (55.4–74.9)
2.20 (1.2–4.2)
0.0082
Previous treatment with bortezomib
77.6% (69.9–84.4)
59.3% (50.8–67.4)
2.38 (1.4–4.0)
0.0005
mPFS, months
3.7 (95% CI 3.0–5.3)
Previous treatment with lenalidomide
67.5% (55.9–77.8)
53.2% (41.5–64.7)
1.83 (0.9–3.5)
0.035
mDOR, months
4.4 (95% CI 3.7–10.8)
n=24 0 Sd (n=122) Sd2
mITT, modified intention-to-treat; OR, odds ratio; ORR, overall response rate; PFS, progression-free survival, Sd, selinexor, dexamethasone. 1. Grosicki S, et al. Lancet. 2020;396:1563-1573; 2. Chari A, et al. N Engl J Med. 2019;381:727-738.
Selective Inhibitors of Nuclear Export (SINEs): Practical Implications Overall response rate to SDd,1 SPd2 and SKd3 in STOMP
Proportion of patients (%)
100
Orally bioavailable4 Available as a monotherapy4 and has been evaluated as a combination therapy3
80 n=4
60 n=11 40
20
Advantages
n=11
n=7
n=7
n=16
n=7
CR VGPR PR
Works in combination therapy to enhance activity of glucocorticoids, including in tumour cells with resistance to glucocorticoids4 A treatment option for patients with penta-refractory disease and with a manageable fiscal impact5 Demonstrated efficacy in patients with high-risk cytogenetic abnormalities and those with renal dysfunction6
Challenges Cytopenias (thrombocytopaenia), gastrointestinal issues (nausea and anorexia) and asthenia are key dose-limiting toxicities7
0 SDd (n=32)
SPd (n=46)
SKd (n=24)
First month is the most challenging time to manage side effects; patients require proactive supportive care to pre-emptively manage side-effect profile8
SDd, selinexor, daratumumab, dexamethasone; SKd, selinexor, carfilzomib, dexamethasone; SPd, selinexor, pomalidomide, dexamethasone; SVd, selinexor, bortezomib, dexamethasone. 1. Gasparetto C, et al. Eur J Haematol. 2021;2:56-65; 2. Chen C, et al. Abstract 653. ASH Annual Meeting 2019; 3. Gasparetto C, et al. Abstract 1366. ASH Annual Meeting 2020; 4. Chen C, et al. Blood. 2018;131:855-863; 5. Bassali J, et al. Clinicoecon Outcomes Res. 2020;12:317-325; 6. Grosicki S, et al. Lancet. 2020;396:1563-1573; 7. Richter J, et al. Ther Adv Hematol. 2020;11:2040620720930629; 8. Mikhael J, et al. Clin Lymphoma Myeloma Leuk. 2020;20:351-357.
Factors Impacting Treatment Choice in RRMM: Drug Resistance and Novel MOAs Emerging treatment options for RRMM
•
•
•
Drug resistance is the main cause of relapse in patients with MM and is associated with an unfavorable prognosis1 As new combinations of antimyeloma drugs are introduced in earlier lines of therapy, patients with RRMM often have disease that is refractory to multiple drugs1 Therefore, drugs with novel mechanisms of action are urgently needed1
Figure from Ramasamy et al., Blood Rev. 2021.2
ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; APN, aminopeptidase N; BCMA, B-cell maturation antigen; BiTEs, bispecific T-cell engagers; Bcl-2, B-cell lymphoma 2; CAR, chimeric antigen receptor; CDC, complement-dependent cytotoxicity; CRBN, cereblon; Iber, iberdomide; mAb, monoclonal antibody; NK, natural killer 1. Pinto V. et al., Cancers (Basel). 2020 Feb 10;12(2):407; 2. Ramasamy et al., Blood Rev. 2021 Feb 9:100808.
147
BELLINI: Study Design • Double blind, randomized 2:1, placebo-controlled phase 3 trial • Primary end point: PFS • Key secondary end points • • • •
ORR ≥VGPR OS QoL/PRO parameters
Patients with RRMM and: • 1-3 lines of prior tx • PI nonrefractory • (N=291) Stratification by: BTZ-sensitive vs BTZ-naïve Prior lines of tx (1 vs 2-3)
Kumar. ASCO 2020. Abstr 8509.
Venetoclax 800 mg qd + BTZ 1.3 mg/m2 + DEX 20 mg (n=194)
Placebo + BTZ 1.3 mg/m2 + DEX 20 mg (n=97)
148
BELLINI: Efficacy Outcome
Ven + Vd (n=194)
Pbo + Vd (n=97)
Significance (95% CI)
Median PFS, mos
23.22
11.41
HR, 0.60 (0.43-0.82) P=0.0013
Median OS, mos
33.5
NR
HR, 1.46 (0.91-2.33) P=0.112
84
70
P=0.013
≥VGPR
63
40
P<0.001
≥CR
29
7
P<0.001
<10–4
21
4
P<0.001
<10–5
15
2
P<0.002
<10–6
8
1
P=0.037
Median DoR, mos
NR
12.8
HR, 0.46 (0.30-0.71) P<0.001
Median TTP, mos
NR
12.2
HR, 0.55 (0.38-0.79) P=0.001
ORR, %
MRD, %
Kumar. ASCO 2020. Abstr 8509.
149
BELLINI: PFS and OS in Key Patient Subgroups Group, HR (95% CI)
PFS
OS
All patients
0.63 (0.44-0.89)
1.46 (0.91-2.33)
ISS staging I II III Cytogenetic risk High Standard t(11;14) status Positive Negative
0.41 (0.24-0.71) 0.91 (0.49-1.69) 1.23 (0.50-3.05)
1.05 (0.49-2.23) 1.58 (0.74-3.38) 2.41 (0.89-8.36)
Group, HR (95% CI) t(4;14) status Positive Negative
0.18 (0.05-0.76) 0.66 (0.45-0.97)
Kumar. ASCO 2020. Abstr 8509.
3.29 (0.92-11.80) 1.10 (0.64-1.89) 0.72 (0.14-3.60) 1.44 (0.86-2.41)
OS
2.40 (0.88-6.55) 0.53 (0.37-0.75)
3.75 (0.79-17.80) 1.22 (0.72-2.04)
0.52 (0.31-0.89) 1.16 (0.39-3.41)
1.47 (0.71-3.04) 4.05 (0.92-17.90)
0.33 (0.18-0.61) 0.80 (0.52-1.22)
0.92 (0.39-2.16) 2.55 (1.23-5.32)
BCL2 expression (IHC)
High Low 1.21 (0.58-2.52) 0.54 (0.36-0.83)
PFS
BCL2 gene expression (qPCR)
High Low
Peptide–Drug Conjugates (PDCs)
PDCs mechanism of action: example of melphalan flufenamide (melflufen)2
1. The PDC readily diffuses through the cell membrane
3. The alkylating payload induces DNA damage to result in cell death
Myeloma cell 2. The PDC binds aminopeptidases; releasing the alkylating payload
Melphalan flufenamide (melflufen) Phase 2, single-arm, multicentre HORIZON Study (n=157)3 Eligibility criteria: • Aged ≥18 years • ECOG performance status of 0–2 • ≥2 prior lines of therapy (including IMiD and PI) • Refractory to pomalidomide and/or an anti-CD38 mAb
Melflufen 40 mg on D 1 + dexamethasone 40 mg on D 1, 8, 15, and 22 of each 28-day cycle All-treated population (n=157)3
Patient response to melflufen in HORIZON3 Proportion of patients (%)
PDCs are composed of three vital components: a homing peptide, a linker and a cytotoxic payload, which work together to target the selected receptor of a tumour cell and deliver cytotoxins1
50 sCR
VGPR
40 30 20 10
n=1 n=17 n=28
n=13
PR
mPFS, months (95% CI)
4.2 (3.4–4.9)
Median duration of ≥PR, months (95% CI)
5.5 (3.9–7.6)
Most common Grade 3–4 TEAEs in all-treated population (n=157)3
n=18
Neutropaenia (79%)
0 All-treated population (n=157)
Triple-class refractory (n=119)
Thrombocytopaenia (76%)
D, day; PDC, peptide–drug conjugate; TEAE, treatment-emergent adverse event. 1. Cooper BM, et al. Chem Soc Rev. 2021;50:1480-1494; 2. Mateos M-V, et al. J Clin Med. 2020;9:3120; 3. Richardson PG, et al. J Clin Oncol. 2021;39:757-767.
Anaemia (43%)
Peptide–Drug Conjugates (PDCs) HORIZON: subgroup analysis in patients with EMD (n=55):1 • Melflufen + dexamethasone showed activity in patients with advanced RRMM with EMD, a population with high unmet medical need, in the HORIZON study
Patients with EMD (n=55)1 ORR, % (95% CI)
24 (13.2–37.0)
mPFS, months (95% CI)
2.9 (2.0–3.7)
DOR (95% CI)
5.5 (1.8–NE)
aPatients
Phase 1/2a, open-label, ANCHOR Study2 Eligibility criteria: • Aged ≥18 years • ECOG performance status 0‒2 • 1–4 prior lines of therapy • Refractory to (or intolerant of) an IMiDa • Not refractory to PI in last line of therapy • Measurable disease
Patient response to melflufen + Dd or 2,3 100 melflufen + Vd in ANCHOR sCR 90 80 CR n=1 70 n=3 VGPR 60 n=2 50 n=10 PR 40 n=3 30 20 n=11 n=4 10 0 Melflufen + Dd Melflufen + Vd (n=33) (n=14) Next steps: Phase 3 LIGHTHOUSE study: melflufen and dexamethasone in combination with daratumumab compared with daratumumab alone4
Proportion of patients (%)
Melphalan flufenamide (melflufen)
Melflufen 40/30/20 mg + daratumumab 16 mg/kg + dexamethasone 40 mgb or melflufen 40/30/20 mg + bortezomib 1.3 mg/m2 + dexamethasone 20/40 mgb
Most common Grade 3–4 TRAEs Melflufen + Dd (n=33)2
Melflufen + Vd (n=14)3
Neutropaenia (58%)
Thrombocytopaenia (79%)
Thrombocytopaenia (55%)
Neutropaenia (50%)
Anaemia (24%)
Anaemia (43%)
Melflufen + Dd (n=33)2
Melflufen + Vd (n=14)3
mPFS, months (95% CI)
11.5 (6.7–NR)
Not mature
mDOR, months (95% CI)
12.5 (8.3–NR)
Not mature
assigned to the daratumumab arm could not have received prior anti-CD38 mAb therapy; patients assigned to the bortezomib arm could not have been PI-refractory; bDexamethasone 40 mg was administered q1w in the daratumumab arm and as 20 mg on D 1, 4, 8, and 11 then as 40 mg on D 15 and 22 in the bortezomib arm. Dd, daratumumab, dexamethasone; EMD, extramedullary disease; NE, not evaluable; NR, not reached; q1w, once weekly; RRMM, relapsed/refractory multiple myeloma; TRAE, treatment-related adverse event. 1. Richardson PG, et al. Abstract 3214. ASH Annual Meeting 2020; 2. Ocio EM, et al. Abstract 417. ASH Annual Meeting 2020; 3. Hájek R, et al. Poster 8037 ASCO Annual Meeting 2021; 4. ClinicalTrials.gov: NCT04649060. Available at: https://clinicaltrials.gov/ct2/show/NCT04649060. Accessed 1 September 2021.
Peptide–Drug Conjugates (PDCs) Melphalan flufenamide (melflufen) Phase 3, randomised, head-to-head, multicentre OCEAN Study (n=495)1,2 Eligibility criteria: • Aged ≥18 years • ECOG performance status of 0‒2 • 2–4 prior lines of therapy (including lenalidomide and a PI) • Refractory to lenalidomide within 18 months of randomisation and last line of therapy
Primary endpoint
mPFS, months
Secondary endpoints
ORR, % (95% CI) CR, % VGPR, % PR, %
Hazard ratio (95% CI)
OS, months Hazard ratio (95% CI)
Melflufen + dexamethasone (n=246)1
Pomalidomide + dexamethasone (n=249)1
6.8
4.9
1:1
Melflufen 40 mg + dexamethasone 40 mg or pomalidomide 4 mg + dexamethasone 40 mg
Most common Grade 3–4 TEAEs1 Melflufen + dexamethasone (n=228)
Pomalidomide + dexamethasone (n=246)
Thrombocytopaenia (76%)
Neutropaenia (49%)
Neutropaenia (64%)
Infection (22%)
Anaemia (43%)
Anaemia (18%)
Infection (13%)
Thrombocytopaenia (13%)
0.79 (0.64–0.98); p=0.03 33 (27–39) 3 9 20
27 (22–33) 1 7 18
19.8
25.0 1.10 (0.85–1.44)
Data cutoff: 3 February 2021. 1. Schjesvold F, et al. Abstract OAB-50 IMW 2021; 2. ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/NCT03151811. Accessed 1 September 2021.
Selected Immune Therapies • CAR T Therapy • Bispecific T Cell Engagers
• MoAb/drug conjugate: GSK 2857916 (“belamaf”) • Emerging role of CelMods (iberdomide, CC92480)
CAR T, chimeric antigen receptor T cells.
Antibody Drug Conjugates (ADC) – BCMA Auristatin Immunotoxin Induces Strong Anti-MM Effects Macrophage (M )
Apoptotic MM cells MMAF released at lysosome to induce G2/M arrest followed by apoptosis
ADPC ADCC FcRII
Apoptosis FcRII
NK , Monocyte M engulfing MM
MM
MM
GSK2857916
NFB
MM cell lysis
Inhibition of NFB signaling
BAFF
APRIL
Bone Marrow Stromal Cell BCMA
GSK2857916
ADCC, Antibody-dependent cellular cytotoxicity; ADCP; antibody-dependent cellular phagocytosis; APRIL, A proliferation-inducing ligand; BAFF; Bcell activating factor; BCMA, BCMA, B-cell maturation antigen; MM, multiple myeloma; Tai YT et al. Blood. 2014;123(20):3128–3138.
Antibody–Drug Conjugates (ADCs) ADCs are composed of an antibody, which binds to a protein highly expressed in tumour cells, connected by a linker to a cytotoxic small-molecule payload, enabling the targeted delivery of the drug to the tumour1 ADCs mechanism of action: example of belantamab mafodotin1,2
Belantamab mafodotin Phase 2, open-label, two-arm, multicentre DREAMM-2 Study (n=196)2 Eligibility criteria: • Aged ≥18 years • ECOG performance status of 0–2 • Disease progression after ≥3 lines of therapy • Refractory to IMiDs, PIs and refractory and/or intolerant to an anti-CD38 mAb
BCMA Anti-BCMA monoclonal antibody
Myeloma cell
Proportion of patients (%)
MMAF
Response to belantamab mafodotin by dose in DREAMM-22 CR/sCR 50
VGPR
40 30 20
n=3
PR
n=3 n=15
10
n=17
n=12
n=14
2.5 mg/kg (n=97)
3.4 mg/kg (n=99)
0
ADC, antibody–drug conjugate; BCMA, B-cell maturation antigen; DOR, duration of response; MMAF, monomethyl auristatin F. 1. Cooper BM, et al. Chem Soc Rev. 2021;50:1480-1494; 2. Lonial S, et al. Lancet Oncol. 2020;21:207-221.
1:1
Belantamab mafodotin • 2.5 mg/kg (n=97) • 3.4 mg/kg (n=99)
2.5 mg/kg (n=97)
3.4 mg/kg (n=99)
mPFS, months (95% CI)
2.9 (2.1–3.7)
4.9 (2.3–6.2)
Probability of DOR of ≥4 months (95% CI)
78% (57–89)
87% (69–95)
Most common Grade 3–4 AEs2 2.5 mg/kg (n=95)
3.4 mg/kg (n=99)
Keratopathy (27%)
Thrombocytopaenia (33%)
Thrombocytopaenia (20%)
Anaemia (25%)
Anaemia (20%)
Keratopathy (21%)
Belantamab Mafodotin in Combination with Pomalidomide and Dex for RRMM With PIs, IMiDs, anti-CD38, BMCA-targeted agents are the 4th pillar of MM treatment BELAMAF is the only anti-BMCA treatment, available for all patients with 30 minute outpatient infusion Bela-Pd demonstrates exceptional efficacy (> VGPR 72%; ORR 98 %) in patients that are IMiD/PI/Dara refractory BELAMAF 1.92 mg/kg Q4W > VGPR 64% and median PFS 14.1 months
Grade 3/4 keratopathy in 25% and < 20/50 BCVA 17% BELAMAF 2.5 mg/kg (SINGLE Loading, SPLIT) > VGPR 74% (100% for the 2.5 mg/kg Q4W) and not yet reached
Grade 3/4 keratopathy in 70% and < 20/50 BCVA 15% Alternative dosing schedules are under evaluation to further optimize efficacy/safety profile Trudel et al ASH 2020
Antibody-Drug Conjugates (ADCs): Practical Implications Overall response rate to belantamab mafodotin + Vd1 and belantamab mafodotin + Pd2 in DREAMM-6 100
Proportion of patients (%)
sCR 80
n=4
VGPR
Advantages Off-the-shelf availability for immediate administration in an outpatient setting, particularly important for patients with rapidly progressing disease3,4 Suitable for use in elderly patients, due to lack of any observed cases of CRS or ICANS3,4
Available as a monotherapy5 PR 60 n=9 n=15
Targeted cytotoxicity means it acts independently of T-cell fitness4 Studies of belantamab mafodotin as part of combination regimens are underway1,2,4
40
Challenges 20 n=5
n=6
0 Belamaf + Vd (n=18)
Belamaf + Pd (n=29)
Ocular toxicity is a major concern. Patients receiving belantamab mafodotin must be enrolled in a risk evaluation and mitigation strategy programme and must undergo ophthalmic examinations at baseline and prior to each dose. Patients should be advised to use preservative-free lubricant eye drops and avoid contact lenses during treatment5 Dose range and frequency to minimise side effects are being evaluated (e.g. longer infusion times and dosing delays)6
CRS, cytokine-release syndrome; ICANS, immune effector cell-associated neurotoxicity syndrome; Pd, pomalidomide, dexamethasone. 1. Popat R, et al. Abstract 1419. ASH Annual Meeting 2020; 2. Trudel S, et al. Abstract 1419. ASH Annual Meeting 2020; 3. Becnel MR, Lee HC. Ther Adv Hematol. 2020;11:2040620720979813; 4. Offidani M, et al. Drug Des Devel Ther. 2021;15:2401-2415; 5. BLENREP (belantamab mafodotin) [prescribing information]. Brentford, Middlesex, UK: GlaxoSmithKline; 2020; 6. Speaker’s clinical experience.
158
IBER enhances in vitro immune-stimulatory activity versus LEN and POM1 LEN2
IL-2 secretion by treated PBMCs3
IBER2
Compound concentration required for degradation of Ikaros or Aiolos protein in vivo:2 EC50, nM2
Ikaros
120
LEN POM IBER
LEN POM IBER
100
Live cells (% DMSO)
Secreted IL-2 (ng/mL)
1500
MM cell survival in co-culture with treated PBMCs3
1000
500
80
60
40
Aiolos
LEN
67
87
POM
24
22
IBER
1
0.5
0 20 0
0.1
1
10
100 1,000 100,000
Compound concentration (nM)
0
0.1
1
10
100 1,000 100,000
Compound concentration (nM)
Investigational only, not approved. Figure extracted from Matyskiela ME, et al. J Med Chem. 2018;61:535-42. DMSO, dimethylsulfoxide; EC50, half maximal effective concentration; IBER, iberdomide; IL, interleukin; LEN, lenalidomide; MM, multiple myeloma; PBMC, peripheral blood mononuclear cell; POM, pomalidomide. 1. Bjorklund CC, et al. Leukemia 2020;34:1197–201. 2. Matyskiela ME, et al. J Med Chem. 2018;61:535-42. 3. Lonial S, et al. Presented at ASCO 2019; abstract 8006.
Cereblon E3 Ligase Modulators (CELMoDs) CELMoDs, a type of IMiD, target cereblon, leading to myeloma cell apoptosis and immune system stimulation1 CELMoDs mechanism of action: Example of Iberdomide (CC-220)2 Proteasome
Iberdomide (CC-220) Phase 1/2, dose-escalation CC-220-MM-001 study (n=40)3 Eligibility criteria: • Aged ≥18 yearsa • Prior therapies (including lenalidomide and/or pomalidomide, and a PI): – ≥2 for IberDd cohort – ≥1 for IberVd cohort • PD on or within 60 days of last therapy Patient response to IberDd and IberVd in CC-220-MM-0013
Cul4A
RBX1
DDB1 CRBN
Ub
CC-220
Substrate
Substrate degradation
aIn
Proportion of patients (%)
Substrate
Most common Grade 3–4 TEAEs of interest3
50 40 30
n=1
CR
n=3
VGPR PR
n=2
20 10
n=4
n=6
IberDd (n=19) or IberVd (n=21)
IberDd (n=19)
IberVd (n=21)
Neutropaenia (50%)
Neutropaenia (20%)
Leukopaenia (22%) Anaemia (22%)
Thrombocytopaeni a (20%)
0 IberDd (n=19)
IberVd (n=21)
•
mDOR was not reached
patients aged <65 years, presence of important comorbid conditions(s) likely to have a negative impact on tolerability of high-dose chemotherapy with ASCT was required for inclusion. CRBN, cereblon; Cul4A, cullin 4A; DDB1, damage-specific DNA binding protein 1; IberDd, iberdomide, daratumumab, dexamethasone; IberVd, iberdomide, bortezomib, dexamethasone; PD, progressive disease; RBX1, RING-box protein 1; Ub, ubiquitin. 1. Bobin A, et al. Cancers (Basel). 2020;12:2885; 2. Gao S, et al. Biomark Res. 2020;8:2; 3. Van De Donk NWCJ, et al. Abstract 724. ASH Annual Meeting 2020.
Cereblon E3 Ligase Modulators (CELMoDs): Practical Implications
100
Patient response to iberdomide 0.3‒1.2 mg + dexmethasone1
Potential advantages
Proportion of patients (%)
VGPR
Higher potency than previous IMiDs3
80 PR 60
40
Possibility to overcome resistance to certain drugs2,3
Efficacy shown in heavily pre-treated patients in combination regimens1,4 Promising clinical activity among patients refractory to the last regimen and previously exposed to IMiDs, PIs and CD38 mAbs4
n=1
20 n=15
Potential challenges Understanding the relationship between mutational status and clinical response; CELMoDs may be ineffective in cases where IMiD resistance results from CRL4CRBN component or Ikaros mutations3
0 Iberdomide + dexamethasone (n=51)
CRL, cullin-RING E3 ubiquitin ligase; MR, minimal response; SD, stable disease. 1. Lonial S, et al. Abstract 8006. ASCO Annual Meeting 2019; 2. Bobin A, et al. Cancers (Basel). 2020;12:2885; 3. Davis LN, Sherbenou DW. Cancers. 2021;13:1686; 4. Van De Donk NWCJ, et al. Abstract 724. ASH Annual Meeting 2020.
161
CC-92480 is a novel CELMoD® agent specifically designed for rapid protein degradation1,2 Efficient substrate degradation leading to apoptosis and potent antiproliferative activity in LEN and POM resistance3 Aiolos degradation efficiency1
Apoptosis induction kinetics
100
7.5
LEN
40
Ymin = 35 Ymin = 18 CC-92480
1 10-6
1
Compound concentration (M)
CC-92480, a potent CELMoD agent1
aDF15R. bDF15,
0.1 µM POM
5.0
2.5
Ymin = 5 0.001
0
0
50
LEN
10,000
Proliferation IC50 (nM)
POM
Caspase-3 induction
Control (%)
60
0
LEN-resistant cells (H929-1051) 0.01 µM CC-92480
80
20
Antiproliferative activity in LEN/POM resistance
100
150
POM
CC-92480
1,000 100
10 1
No cereblona
0.1
Parental cell lineb Resistant cell linec
0.01
Time (hours)
LEN1
POM1
H929, and OPM-2. cH929R1, H929R2, OPM-2R1, OPM-2R2, and OPM-2R3. CELMoD, cereblon E3 ligase modulator; LEN, lenalidomide; POM,
pomalidomide. 1. Hansen J, et al. J Med Chem 2020;63:6648–6676. 2. Wong L, et al. Blood 2019;134:1815. 3. Richardson PG, et al. Presented at ASCO 2020; abstract 8500.
162
Response, n (%)
CC-92480: Best Response
•
100 80 60 40 20 0
CBR 26.3%
ORR 21.1% 1 (1.3) 6 (7.9) 9 (11.8) 4 (5.3) 37 (48.7)
CBR 50.0%
SD or better 75.0%
ORR 40.0%
ORR 54.5%
2 (20.0)
1 (9.1) 2 (18.2)
2 (20.0) 1 (10.0)
CBR 63.6% SD or better 100%
5 (50.0) 15 (19.7)
All Evaluable (n = 76)
3 (27.3)
DCR 100%
1 (9.1) 4 (36.4)
3 (3.9)
10/14 days x 2 1.0 mg QD (n = 10) MTD
CR VGPR PR MR SD PD NE
21/28 days 1.0 mg QD (n = 11) RP2D
7 of 11 patients at the RP2D of 1 mg QD 21/28 days were triple-class refractory (to ≥ IMID, 1 PI, and 1 anti-CD38 mAb) •
Of these patients, 1 had CR, 1 VGPR, 2 PR, and 1 MR Richardson PG et al, ASCO 2020. Abstr 8500.
163
Responses in patients with EMD • Only patients on continuous schedules are shown Dosing Dose level schedulea
C2
C3
0.1 mg QD 0.2 mg QD
SD PD
PD
10/14 days × 2
0.3 mg QD 0.6 mg QD
21/28 days
10/14 days × 2
C4
C5
C6
C7
C8
1.0 mg QD
SD PD PD SD SD
VGPRa
PD
PRb MR
PD
SD PDc
SD PR SD MR SD
On treatment at time of data cut
PD
PR SD
1.0 mg QD
PD SD PR VGPR PR VGPR PR (case study) SD
PET scan post-CC-92480 C3D1
VGPR PR
PR
21/28 days
C10
CR
SD
0.8 mg QD
C9
PET scan pretreatment
CR
1.0 mg dose active in EMP
a1
patient in the 21/28-day 1.0 mg QD cohort had an unconfirmed VGPR as of the data cutoff date; b1 patient in the 21/28-day 0.8 mg QD cohort had an unconfirmed PR as of the data cutoff date; c1 patient in the 21/28-day 0.8 mg QD cohort had an unconfirmed PD as of the data cutoff date. C, Cycle; CR, complete response; D, Day; EMP, extramedullary plasmacytoma; MR, minimal response; PD, progressive disease; PET, positron emission tomography; PR, partial response; QD, once daily; SD, stable disease; VGPR, very good partial response .
Richardson PG, et al. Presented at ASCO 2020; abstract 8500.
Conclusions ~ Integration and Impact of Novel Agent Combinations in RRMM, including Immune Therapies, and Overcoming Key Challenges… •
Innovations (PIs, IMiDs, mAbs, HDACi’s) to date have produced significant improvements in PFS and OS; recent approvals will augment this – with Quad Combinations emerging as a new standard
•
The treatment landscape for RRMM is ever changing as new therapies are being developed, offering hope to patients
•
Further investigation and clinical practice in the real world will determine how these drugs fit into the treatment landscape for RRMM, through optimisation of dosing and AE management
• • •
Next wave of immune therapies: crucially, are they agnostic to mutational thrust? Baseline immune function is a key barrier to success and is targetable mAbs (including ADCs, BiTEs) represent true new novel mechanisms, as well as other immunotherapeutics (e.g. CAR –T, cellular therapies, vaccines) New insights to mechanisms of drug action are further expanding treatment/immuno-therapeutic opportunities with combinations Next generation small molecules and targeted therapy show great promise – e.g. VENETOCLAX, SELINEXOR and MELFLUFEN, and for new agents in development, e.g. CeLMoDS Further refinement of prognostics, immune profiling, and MRD will guide therapy
• • •
The Impact Of Novel Therapies and the Importance of Sequence in RR MM ~ 2021 2009 – Patient DG, age 62 years High Risk IgG kappa MM DSS 3, ISS 2, Elevated LDH 17 del positive , R-ISS 3 13 del positive (by FISH)
2017
2018
PMH – HTN, requiring triple therapy
RD + Zoledronic acid => RVD (VGPR)
Well tolerated, minimal PN (G1)
2010 ASCT (CY – HDM) (CR) R/Z maintenance 2011 PD – RVD (PR)
2019
2012 PD – Pom VD (VGPR) 2013 PD (aggressive relapse with extra-medullary disease) DARA [501] 16 mg/kg (CR; MRD -) to present ( > 8 years) with multiple future options when needed….now aged 74 years and is a grandmother X3.
“Best I have ever felt since prior to diagnosis, and even despite dealing with the COVID pandemic” Clinic visit @ DFCI 2021
NEJM, 2015
Ongoing MM Collaborative Model for Rapid Translation From Bench to Bedside
Thank YOU! Shawna Corman Investigatorship
Dana and Rob Smith Family Fund For MM Research RJ Corman MM Research Fund
Pharmaceuticals Advocacy
Academia
Progress and Hope NIH NCI
IMF
Paula and Rodger Riney Foundation
MMRF/C IMWG, LLS
FDA EMEA
Counter Myeloma Mike Sheehan
15 Novel Drugs and >30 new FDA-approved drugs/combos/indications in last 18 yrs
Panel Discussion
167
Audience Q&A
• Open the Q&A window, allowing you to ask questions to the host and panelists. They can either reply to you via text in the Q&A window or answer your question live. • If the host answers live, you may see a notification in the Q&A window.
• If the host replies via the Q&A box – you will see a reply in the Q&A window.
IMF Patient and Family Webinar
Webinar Survey & Closing Remarks 169
We Want to Hear From You! Feedback Survey At the close of the meeting a feedback survey will pop up. This will also be emailed to you shortly after the workshop. Please take a moment to complete this survey.
Thank you to our sponsors!
171
172