2025 IMWG Summit Slides Tuesday

Page 1


WELCOME & INTRODUCTION

S. Vincent Rajkumar, MD

Mayo Clinic, Rochester

imwg.org

imwg.net

imwg.info

imwg.online

imwg.ai

IMWG Website

• Bone Disease Committee (Terpos/Hillengass)

• Immunotherapy Committee (Martin/Lin)

• Mass Spectrometry Committee (Murray)

• SMM Committee (Mateos/Kumar)

• MRD Committee (Paiva/Munshi)

• QOL/PRO Committee (Zweegman/Sidana)

Publications in 2024

Publications in 2024

IMWG Guidelines for Newly Diagnosed Myeloma

IMWG Consensus on Solitary Plasmacytoma

IMWG Guidelines for Smoldering Myeloma

IMWG Recommendations for Dose and Schedule of Myeloma

Drugs and Regimens

Immunotherapy

Thank you to

2025 IMWG Summit Sponsors

SESSION 1:

MGUS AND SMOLDERING MULTIPLE MYELOMA (SMM)

Session Chair: S. Vincent Rajkumar, MD Mayo Clinic, Rochester

PROGRESSION

OF MGUS TO MULTIPLE MYELOMA

Sloan Kettering Cancer Center

Memorial

Progression of MGUS to Myeloma

Francesco Maura, M.D. Memorial Sloan Kettering Cancer Center, NY Twitter: @FrancescoMaura4 email: mauraf@mskcc.org

Multiple myeloma pathogenetic model

Multiple myeloma genomic landscape

~2000 patients with WES/targeted sequencing data

>150 genomic drivers

Maura F*, Rajanna A*, Ziccheddu B*, Poos A.*, et al. JCO 2024
Sheri Skerget et al. Nat Gen 2024; Alberge JB et al. Nat Gen 2025
Maura F*, Bolli N* et al Nat Comm 2019
Ziccheddu

Multiple myeloma pathogenetic model

Multiple myeloma pathogenetic model

RAS mutations

APOBEC in myeloma precursor conditions

SBS signatures in normal B- and T-cells

MPC: myeloma precursor conditions

APOBEC is not detectable in stable myeloma precursor conditions and normal B-cells

Oben B. et al. Nature Comm 2021 Machado H. et al.

Timing of MM initiation

Age of first chromosomal gain to age of sample collection was median 30 years.

Heidelberg
Weinhold Raab Poos
Mayo Clinic
Baughn Kumar Rajkumar
Maura F et al. Nat Gen 2025 in press; Rustad et al. Nat Comm 2020; Oben et al. Nat Comm 2021; Maura et al CCR 2021; Samur et al. Blood 2024;

Genomic landscape of multiple myeloma precursor conditions

Myeloma

Bolli et al. Nat Comm 2018; Maura et al. CCR 2020; Machado et al. Nature 2022; Oben et al. Nat Comm 2021; Dutta A.K. et al; Cancer Discov 2023

SMM and MGUS pooled analysis

• 374 patients

• 301 with no early intervention

Absence of myeloma genomic defining events

Malignant transformation in multiple myeloma

Genomic IMWG 2020

How to leverage/maximize immune control

MSKCC

Saad Usmani

Ross Firestone

Juan Jose Garces

Kylee H Maclachlan

Andrew Mcpherson

Andriy Derkach

Alexander Lesokhin

Malin L. Hultcrantz

Urvi Shah

Heather Landau

Ahmet Dogan

Yanmin Zhang

Misha Roshal

Sham Malinakody

Sohrab Shah

Moffit Cancer Center

Ken Shain

Fred Locke

Ciara Freeman

Ariosto Silva

Marilena Tauro

Mark B Meads

Mike Jain

Danny De Avila

Praneeth Reddy Sudalagunta

Rafael Renatino Canevarolo

Erin M. Siegel

Phaedra Agius

Jamie Teer

Rachid Baz

Doris Hansen

Leif Begsagel

Marta Chesi

Esteban Braggio

Erin Wiedmeier-Nutor

Linda Baughn

Yusuke Yamashita

Yan Asmann

Shaji Kumar

Rafael Fonseca

Vincent Rajkumar

MDACC

Robert Orlwoski

Krina Patel

Sanger Institute

Peter Campell

Daniel Leongamornlert

UM – SCCC

Ola Landgren

Bachisio Ziccheddu

Benjamin Diamond

Marios Papadimitrioui

Michael Durante

University of Calgary

Nizar Bahlis

Paola Neri

Holly Lee

Mansour Poorebrahim

And many many more….

Heidelberg & GMMG

Niels Weinhold

Alexandra Poos

Elias K. Mai

Hartmut Goldschmidt

Katja C. Weisel

Roland Fenk

Marc-Andrea Bärtsch

Marc S. Raab

NYU

Gareth Morgan

Patrick Blaney

University of Milan

Niccolò Bolli

Matteo Da Via

Giancarlo Castellano

Alessio Marella

Akihiro Maeda

Marta Lionetti

Antonio Matera

Stefania Pioggia

Matteo Claudio Da Vià

Claudio de Magistris

Hasselt University

Benedith Oben

Guy Froyen

University of Würzburg

Leo Rasche

Hermann Einsele

Twitter/X: @FrancescoMaura4

Email: mauraf@mskcc.org

RISK STRATIFICATION AND MANAGEMENT OF SMM

Meletios A. Dimopoulos, MD

University of Athens School of Medicine

Risk Stratification and Management of SMM

Meletios(Thanos)Dimopoulos, MD

Professor and Chairman

Department

Plasma

National

Disclosures

The 20-2-20 model

• median follow-up: ~6.2 years

• 29% cumulative progression rate at 2 years

• Single center

• n = 421 patients, with SMM diagnosis in 2003-2015

• Excluded those with SLiM (retrospectively)

• Advanced imaging in 124 (29.4%)

Risk factors

• median follow-up: 3 years

• 22% cumulative progression rate at 3 years

• Multicenter

• n = 1363 patients, with SMM diagnosis in 20042018

• Excluded those with SLiM

• Advanced imaging in unknown number

How does 20-2-20 perform in modern SMM cohorts ?

median follow-up : 3 years

12% cumulative progression rate at 3 years

• n = 427 SMM patients diagnosed after 2014

• Advanced imaging in 100%

• PD based on SLiM-CRAB criteria

• Majority low risk

• 12% cumulative PD rate at 3 years

• 43% of progressions by SLiM only

• TTP of Low risk SMM ~ MGUS indicating that the current cutoff of 10% BMPC is arbitrary

What is the 20-2-20 risk group distribution and outcome in different SMM cohorts ?

Lakshman Mateos Visram Abdallah Rajkumar

20-2-20 high-risk group outcome in

2-year progression rate among high risk per 20-2-20 according to % of those with advanced imaging in the respective cohort (more advanced imaging = lower progression rate)

Kastritis et al
Abdallah et al
Lakshman et al

New stratification models, beyond 20-2-20

• PANGEA and PANGEA 2.0 models: Trajectories of age, serum creatinine concentration, hemoglobin, paraprotein levels and FLC ratios, with or without BMPC% (Cowan A et al Lancet Haematol 2023 Chabrun F et al ASH 2024)

• Circulating plasma cells (risk stratification and non invasive assessment of evolution): Cutoffs of >0.015% (Terminι R et al) or 0.0014% (Kastritis E et al) (Termini R et al Clin Cancer Res 2022;Kastritis E et al ASH 2024/Submitted)

• Genomic stratification models: Alterations of the MAPK pathway , DNA repair pathway and MYC and APOBEC associated mutations ; distinct genetic subtypes based on mRNA signatures; single cell transcriptional signatures (Bustoros M et al J Clin Oncol. 2020; Bustoros M et al Nat Commun. 2022; Sklavenitis-Pistofidis R et al ASH 2024)

• Immune biomarkers (Bidikian N et al ASH 2024)

Treatment of patients with smoldering MM?

QUIREDEX study

High risk SMM : Len-dex vs observation (n = 119)

Median follow-up: 12.5 years

E3A06 study

Randomized Trial of Lenalidomide vs observation in SMM

Mateos MV, et al. Eur J Cancer 2022

Mateos MV, et al. Lancet Oncology 2016

Mateos MV, et al. NEJM 2013

HR: 0.28 (95%CI 0.120.62)

Median follow-up: ~3 years (A) high risk (B) intermediate risk (C) low risk.

PD: required biochemical PD + by IMWG criteria for MM and evidence of endorgan damage

Lonial S et al J Clin Oncol 2019

Earlier studies in SMM: main limitations

• Disease definition: patients with active MM per IMWG 2014 were included (i.e. BMPCs>60%, FLCr>100)

• Risk stratification was based on older tools

• Non-advanced imaging was used for initial assessments and follow up

• Follow up was less stringent according to current standards

AQUILA study considerations

• SMM defined by current criteria (IMWG 2014)

• High risk criteria were protocol defined (before 2018)

• Central labs every three months

• Advanced imaging used at baseline and every year

• Bone marrow biopsies performed at least every two years

• IMWG criteria were used to define symptomatic MM

Are AQUILA results applicable to current SMM population?

Risk definitions in AQUILA and in RW cohort

AQUILA: Progression to MM by IMWG SLiM-CRAB Criteria (IRC Assessment)

• DARA significantly reduced the risk of progression to MM or death by 51% versus active monitoring for all subsets and risk categories, more pronounced in high risk SMM

• The benefit continued beyond 36 months

• May 20, 2025: FDA ODAC positive vote

Time to MM according to Risk definitions in AQUILA in a RW cohort

N=427 patients diagnosed between 2014-2023, median follow up: 3 years M-protein <3 vs >3 gr/dl FLC ratio <8 vs >8-<100

All SMM patients

Source: Department of Clinical Therapeutics ; Kastritis E et al Blood Adv 2025

What is the natural history of SMM in RW (without treatment) today?

AQUILA: Progression events to MM by IMWG SLiM-CRAB Criteria (IRC Assessment)

Patterns of progression among 427 Smoldering Myeloma patients diagnosed after 2014: importance of monitoring

With modern criteria, use of advanced imaging at baseline and follow-up, the majority of patients are detected by SLiM criteria or asymptomatic bone lesions (no bone fractures)

Which

treatment for high-risk SMM?

Lenalidomide

• Data from small studies

• SMM populations not meeting current definitions

• High risk SMM was minority

• PO administration

• Single agent activity ~55% ORR

• High discontinuation rate (~30->40%)

• Slightly increased risk of second primary malignancies

• Low cost

Daratumumab

• Data from large study

• SMM populations per current definitions

• High risk SMM was minority

• SC administration

• Single agent activity ~64% ORR

• Low discontinuation rate (<5%)

• No increased risk of second primary malignancies

• High cost

SMM treatment strategies: considerations

Delaying disease progression

• Randomized trials available

• No cure

• Fixed duration treatment

• Low toxicity

• Many and increasing salvage options

• Low/high costs

• Applicable to elderly/frail

Curative strategy

• Only phase 2 studies so far

• Some patients may be cured?

• Fixed duration treatment

• Higher toxicity depending on regimen

• Salvage options may be less, but increasing

• High costs

• Applicable to younger/fitter

Multi-drug regimens for SMM (Phase 2 studies)

Ongoing studies for SMM

New options for SMM: immunotherapy

Bi-specifics

• Teclistamab monotherapy in high risk SMM (Immuno-PRISM trial, NCT05469893).

• N=12, ORR: 100% ; 42% CR, 25% VGPR, 33% PR.

• Elranatamab in patients with previously untreated high-risk SMM / EMN34 study (NCT06183489)

• Linvoseltamab in SMM (NCT05955508)

• Teclistamab or Talquetamab in Combination With Daratumumab for High-Risk Smoldering Myeloma (REVIVE Study)(NCT06100237)

Cellular therapies

• Cilta-Cel in SMM

• CAR- PRISM trial, NCT05767359)

• CAR-HiRiSMM tria, NCT06574126

• A Study of Autologous Expanded Natural Killer Cell Therapy for Asymptomatic Multiple Myeloma (UARK 2013-05, NCT01884688

SMM: where are we today?

• We now have four options for HR SMM

• Early intervention with daratumumab (when approved)

• Close follow up with labs (Q 3-4 months) and modern imaging techniques (Q year)

• Enrollment in trials with combinations of current agents active in MM (PI,IMiDs,antiCD38)

• Enrollment in trials with curable intent (HDT, CARs, Bispecifics)

• Decisions may be based on patient’s wish, fitness/frailty, comorbidities, life expectancy

• No need for trials in low risk SMM

• For intermediate risk SMM: we need trials with innovative approaches to intercept evolution to myeloma

• Research to identify risk factors at baseline that may move patients to an early myeloma category

• Research to develop dynamic models which are more informative for the daily management of patients (evolving patterns)

National and Kapodistrian University of Athens

Department of Clinical Therapeutics

(HORIZON-MISS-2021-CANCER-02-03)

Elucidation of risk factors and health determinants associated with progression of monoclonal gammopathies to multiple myeloma

(HORIZON-MISS-2021-CANCER-0201)

Early detection and screening of hematological malignancies

PANEL DISCUSSION

Panelists:

María-Victoria Mateos, MD, PhD

University of Salamanca &

Irene Ghobrial, MD

Dana-Farber Cancer Institute

SESSION 2: RISK AND RESPONSE ASSESSMENT

Session Chair: Nikhil Munshi, MD

Dana-Farber Cancer Institute

NEW MYELOMA RISK STRATIFICATION UPDATE

University Cancer Institute of Toulouse

Oncopole

New Myeloma Risk Stratification

The IMS/IMWG consensus definition of high risk myeloma

Unit for Genomics in Myeloma, Toulouse, FRANCE

DISCLOSURES

Honoraria: Janssen, Takeda, Amgen, Sanofi, Bristol Myers Squibb, Pfizer, Adaptive, Abbvie

Consulting or advisory board : None

Research funding: Sanofi, Bristol Myers Squibb

Travel Support: Janssen, Sanofi, Bristol Myers Squibb, Pfizer

Revised International Staging System (R-ISS)

Palumbo et al. J Clin Oncol 2015
Greipp et al. J Clin Oncol 2005

R2-ISS from European Myeloma Network

3340

NDMM patients

D’Agostino et al. J Clin Oncol 2022

Workshop on “Genomics Defining High-Risk Myeloma”

Barcelona July 7-8, 2023

in more than 20% of sorted plasma cells

on genomic definition of high risk myeloma

Association of 2 among t(4;14) or t(14;16) or t(14;20)

IMS/IMWG consensus on genomic definition of high risk myeloma

Del17p in more than 20% of sorted plasma cells

TP53 mut (no threshold VAF)

Biallelic Del(1p32)

Association of 2 among t(4;14) or t(14;16) or t(14;20)

Gain/Amp 1q Monoallelic del(1p32)

Avet-Loiseau et al. J Clin Oncol, in

Revised International Staging System (R-ISS)

Del17p ISS stage I/II are NOT defined high risk

Palumbo et al. J Clin Oncol 2015
Greipp et al. J Clin Oncol 2005

ISS in 877 NDMM patients with del(17p)

Unpublished data

Heterogeneity of intermediate risk subroup (R-ISS II)

ISS in 877 NDMM patients with del(17p)

Unpublished data

Heterogeneity of intermediate risk subroup (R-ISS II) No del17p Del17p

Del17p : which prognostic cut off by FISH ?

• R-ISS : « higher that the cutoff threshold defined by each lab »

• R2-ISS : 10 to 20%

• IFM : 50-55%

• Until 1% in some clinical studies !

Del17p : which prognostic cut off by FISH ?

OS according to del17p

>1000 del(17p) patients

Del(17p) 0-29%

Del(17p) 30-49%

Del(17p) 50-100%

Thakurta et al. Blood 2019

(36.0m)
HARMONY dataset

Del17p : which prognostic cut off by FISH ?

del17pconfershighriskfrom20%

IMS/IMWG consensus on genomic definition of high risk myeloma

Del17p in more than 20% of sorted plasma cells

Association of 2 among t(4;14) or t(14;16) or t(14;20)

Gain/Amp 1q Monoallelic del(1p32)

Avet-Loiseau et al. J Clin Oncol, in

Isolated del17p
Double hit

n=34 n=25 n=43 n=541 Do we also need TP53 mutational status ?

Do we also need TP53 mutational status ?

IsolatedTP53mutationconfershighrisk

n=34 n=25 n=43 n=541

IMS/IMWG consensus on genomic definition of high risk myeloma

Del17p in more than 20% of sorted plasma cells

TP53 mut (no threshold VAF)

Biallelic Del(1p32)

Association of 2 among t(4;14) or t(14;16) or t(14;20)

Gain/Amp 1q Monoallelic del(1p32)

Avet-Loiseau et al. J Clin Oncol, in

Prognostic impact of del(1p32) : 10% of NDMM, including 1/5 biallelic

PFS

Prognostic impact of del(1p32) : 10% of NDMM, including 1/5 biallelic

PFS

IMS/IMWG consensus on genomic definition of high risk myeloma

Del17p in more than 20% of sorted plasma cells

TP53 mut (no threshold VAF)

Biallelic Del(1p32)

Association of 2 among t(4;14) or t(14;16) or t(14;20)

Gain/Amp 1q

Monoallelic del(1p32)

Avet-Loiseau et al. J Clin Oncol, in

Heterogeneous prognostic impact of t(4;14)

WT

t(4;14) only

t(4;14) with gain 1q or del1p32

WT

t(4;14) only

t(4;14) with gain 1q or del1p32

t(4;14)=highriskifassociatedtoanotherintermediatelesion (1qgain,del1p32)

IMS/IMWG consensus on genomic definition of high risk myeloma « CONSENSUS GENOMIC STAGING » (CGS)

in more than 20% of sorted plasma cells

Association of 2 among t(4;14) or t(14;16) or t(14;20)

without Renal Failure (Creatinine <1.2 mg/dL)

FORTE study

COMPASSSM dataset

IMS/IMWG consensus on high risk myeloma definition

in more than 20% of sorted plasma cells

2 among t(4;14) or t(14;16) or t(14;20)

Conclusion

• The IMS Panel recommends the use of this HRMM definition in all clinical trials going forward and in routine practice.

• The HR subset should represent around 20% of NDMM patients.

• Represents an important step toward risk-stratified therapeutic approaches in routine.

• We hope that this definition will promote the design and conduct of clinical trials FOCUSED on patients with HRMM.

• NGS-based definition, but available data with FISH (report the cut-offs positivity used).

• Risk features should be also evaluated at relapse* and prior to participation in a clinical trial.

(*the risk status at relapse supplants risk status at diagnosis, unless the patient was deemed high-risk at diagnosis).

• Do we need an ultra high risk definition ?

IMS/ IMWG

definition

t(8;14)

Chromothripsis

t(4;14) breakpoint

t(x;22) (light chains) Immune profile

Number and size of PET or MRI lesions at diagnosis Extra medullary disease

Functional Risk Dynamic response to therapy - MRD

THANKS

Team Genomic and immunology of myeloma

Ludovic Martinet

Aurore Perrot

Anaïs Schavgoulidze

Laure Derrier

Sabrina Maheo

Céline Mazzotti

Luka Pavageau

Chloé Cerutti

Antoine Graffeuil

Coralie Franck

Lesluyes Tom

Nadège Carrié-Constantin

Marie-Véronique Joubert

Liliana Lucca

Pierre-Paul Arixa

Marine Cuisinier

Cazes Amélie

Daunes Hélène

Unit for Genomics in Myeloma

Charlotte Théral

Sandy Bernou

Laurine Martin

Marie Cortez

Marie-Anne Marsili

Jennifer Amassi

Marie-Sarah Mascaraque

Lou-anne Larichaudy

Stéphanie Laffaure

Tambi Ralamboarivony

Emmanuelle Le Trionnaire

Charlotte Avet-Loiseau

Aurélie Berberian

Sophie Renaudin

Jean-Michel Herrera

Nikhil Munshi

Mehmet Kemal Samur

Anil Aktas-Samur

General Considerations for the IMS Consensus HRMM Definition 2024

Feature/Method Consideration(s)

NGS-based methods

iFISH

Sample source/analyte

WGS/targeted NGS should now be used for broad molecular profiling, similar to other hematological malignancies.

 iFISH is not sufficient to fully risk-stratify MM patients.

 If performed, selected/enriched plasma cells, with ≥70% purity, should be used.

 Percentage of cells with abnormalities reported should be corrected based on purity of the sample.

 Mutations in p53 and a small deletion in 1p requires sequencing and iFISH alone is no longer sufficient to detect these changes.

Although data suggest that the molecular profile (CAs, mutations, other genomic alterations) is spatially heterogeneous (i.e., different tumor sites can have different molecular profiles or clonal distributions), currently, bone marrow should be used for assessing the molecular profile.

Timing of risk assessment and risk status definition

It is important to perform risk assessment at diagnosis and at relapse.

 Risk features should be evaluated at relapse and prior to participation in a clinical trial.

 The risk status at relapse supplants risk status at diagnosis, unless the patient was deemed high-risk at diagnosis.

CA, chromosomal abnormality; GEP, gene expression profiling; iFISH, interphase fluorescence in situ hybridization; MM, multiple myeloma

Prognostic heterogeneity of t(4;14) : a matter of NSD2 breakpoint position

(Nuclear Receptor SET Domain Protein 2 = histone methyl transferase)

75,1 months (≈ 45%)

59,4 months (≈ 25%)

28,6 months (≈ 30%)

p=0.00019

Which technique to assess genomic risk in MM ?

FISH (gold standard) Next Generation Sequencing

20 000 plasma cells / probe [cryostor]

≥ 45 000 plasma cells [dry pellet]

From Pawlin C. and Davies F., Blood 2019

Detection of genomic abnormalities by NGS panel

Targeted sequencing panel (3Mb) including :

≈200x depth

- ≈ 2400 SNPs : ALL COPY NUMBER VARIATIONS = del(17p), gain 1q, del(1p32)...

- all IGH sequence: 14q32 TRANSLOCATIONS = t(11;14), t(4;14), t(14;16), t(14;20), t(6;14), t(8;14)

- MUTATIONAL PROFIL

- TP53, NRAS, KRAS, BRAF, FAM46C, DIS3, ATM, ATR, MYC, TRAF3, BIRC2, BIRC3, CYLD, IRF4, CDKN2C…

- CRBN, BCMA, GPRC5D, FCRH5

Detection

of genomic abnormalities by NGS panel

Double focal deletion of CDKN2C (1p32)

Not detected by FISH

Double focal TP53 deletion Not detected by FISH

Focal TP53 deletion

Not detected by FISH

Only at diagnosis ?

 Risk features should be evaluated at relapse and prior to participation in a clinical trial.

 The risk status at relapse supplants risk status at diagnosis, unless the patient was deemed high-risk at diagnosis.

Gain 1q vs Amplification 1q

Niels Weinhold et al., Haematologica 2021

amp(1q) vs. gain(1q) vs. normal 1q: PFS

amp(1q) vs.

Gain 1q vs Amplification 1q

FORTE trial

amp(1q) vs. normal 1q: HR 4.40, 95% CI 2.59–7.49, p<0.0001 amp(1q) vs. gain(1q): HR 3.22, 95% CI 1.89–5.49, p<0.0001 gain(1q) vs. normal 1q: HR 1.37, 95% CI 0.81–2.32, p=0.25

Gain 1q vs Amplification 1q

FORTE trial

Prognostic impact of t(14;16) : 1-3% of NDMM

Schavgoulidze et al. Blood Cancer

1635 NDMM patients

Multivariate analysis of all recurrent abnormalities → Construction and validation of a prognostic model

6 independent variables weighted by a specific coefficient:

• Trisomy 5  - 0.3

• Trisomy 21  0.3

• t(4;14)  0.4

• gain 1q  0.5

• del(1p32)  0.8

• del(17p)  1.2

Score ≤ 0: LOW RISK (≈55%)

Score < 0 & ≤ 1: INTERMEDIATE RISK (≈35%)

Score > 1: HIGH RISK (≈15%)

trisomy 21 trisomy 5 t(4;14) t(14;16)

t(4;14), t(14;16), t(14;20)

Mayo Additive Staging System (MASS)

1327 ND MM patients

et al. Blood Cancer J 2022

Abdallah

NEW RESPONSE CRITERIA UPDATE

Shaji Kumar, MD Mayo Clinic, Rochester

Response Assessment in Myeloma

Malignancies

Scottsdale, Arizona

Rochester, Minnesota

Jacksonville, Florida

DISCLOSURES

• Research funding for clinical trials to the institution: Celgene, Takeda, Janssen, BMS, KITE, Merck, Abbvie, Medimmune, Novartis, RocheGenentech, Amgen, Tenebio, Carsgen

• Consulting/Advisory Board participation: (with no personal payments) Celgene, Takeda, Janssen, Abbvie, Genentech, Amgen, Molecular Partners and (with personal payment) Oncopeptides, Genecentrix, Cellectar.

Why do we need a response criteria?

• Response assessment is critical to assess treatment efficacy

• Uniform assessment allows for comparison of treatment approaches

• Response depth has prognostic value

• Increasingly response depth allows us to adapt treatment approaches

Serum, Urine

Protein

Marrow, Blood, Extramedullary

Measurable Disease

• SPEP ≥ 1g/dl (≥0.5 allowed in relapsed disease)

• Serum iFLC ≥10 mg/dL with an abnormal ratio

• 24-hour urine UPEP ≥200 mg/24 hours can be used in place of FLC assay if not available

• If selected should be followed for response

If none of the above, then -

• BMPC≥30%

• Soft tissue mass (para-skeletal or EMD) with one dimension

≥2 cm

Response categories

• Stable disease - Not meeting criteria for complete response, very good partial response, partial response, or progressive disease

• Nor recommended for use as an indicator of response; stability of disease is best described by providing the time-toprogression estimates.

• Minor Response - ≥25% but ≤49% reduction of serum M-protein and reduction in 24-h urine M-protein by 50–89%.

Response categories

• Partial Response

• If serum is measurable by SPEP - ≥50% reduction of serum M-protein

• If baseline Serum M spike was ≥0.5 and <1, then a reduction to <0.3 gm/dL

• If serum FLC is measurable - ≥50% decrease in the difference between involved and uninvolved FLC levels

• If 24 h urinary M-protein is used as measurable disease, a reduction by ≥90% or to <200 mg per 24 h

• If serum and urine M-protein and serum-free light assay are unmeasurable, ≥50% reduction in plasma cells (provided baseline PC% is > 30%)

Response categories

• Very Good Partial Response (VGPR)

• If serum is measurable by SPEP - ≥90% reduction of serum Mprotein or immunofixation positive only

• If baseline Serum M spike was ≥0.5 and <1, then a reduction to 0 gm/dL

• If serum FLC is measurable - ≥90% decrease in the difference between involved and uninvolved FLC levels or immunofixation positive only

• If 24 h urinary M-protein is used as measurable disease, a reduction to <100 mg per 24 h or immunofixation positive only

Response Categories

• Complete Response

• Negative immunofixation on the serum

• normal FLC ratio or iFLC < ULN

• Negative immunofixation on the 24-hour urine

• and <5% plasma cells in bone marrow aspirates

• sCR (stringent Complete response)

• Complete response as above plus normal FLC ratio iFLC < ULN and absence of clonal cells in bone marrow

* Bone marrow results from within 3 months of the M protein/FLC assessment can be included for CR assessment

Imaging based response

Imaging principles

• Recommendation for functional imaging (PET/CT or DWI-WBMRI) and move away from SPD

• For PSD/EMD functional imaging mandatory to allow response evaluation. MRI preferred for CNS.

• Same technique recommended pre and post therapy.

• Measures of the lesion(s) needed only if no other disease parameters to be quantified available

• 3 months window (+ or -) allowed to match with other tests (BM/PB)

Imaging response categories (with

no plasmacytomas)

PET/CT/DWI-

MRI response Definition

CR (MRD)

PR

SD

PD

Uptake ≤ liver pool (Deauville scale < 4) by PET or RAC-1 by DWI-MRI in all locations (Marrow and focal lesions)

Decrease of either number of focal lesions + stable SUV (DS 4-5),

OR Decrease in activity, with stable number of focal lesions, OR both or RAC-2 on MRI, compared to baseline

No significant change of BM/FL FDG uptake or at MRI RAC-3 compared with baseline

New lesion (FL/EMD/PSD) compared with baseline imaging, both in the functional imaging (PET DS > 4 or MRI RAC 4 or 5) or on CT

Imaging response categories

(with plasmacytomas: paraskeletal or extramedullary)

PET/CT/DWI-

MRI response Definition

CR (MRD)

PR

SD

PD

Uptake ≤ liver pool (Deauville scale < 4) by PET or RAC-1 by DWI-MRI in all locations (BM/FL/PSD/EMD) irrespective of the reduction of soft-tissue plasmacytomas size

Decrease of either number of FLs/PSD/EMD + stable SUV (DS 4-5),

OR Decrease in activity, with stable number of FLs/PMD/EMD, OR both or RAC-2 on MRI, compared to baseline irrespective of the reduction of soft-tissue plasmacytomas size.

No significant change in BM/FL/EMD/PSD uptake or at MRI (RAC-3) compared with baseline, irrespective of soft-tissue plasmacytoma size

New lesion (FL/EMD/PSD) compared with baseline imaging, both in the functional part (PET DS > 4 or MRI RAC 4 or 5) or on CT

First evaluation after 3 months from the start of therapy (to avoid as much as possible background influence/tumor flare after CART/ bone regeneration). If PR/SD: repeat imaging every 3-6 months until CMR/RAC-1. Once CR established, no other evaluation requested until suspect of progression

Minimal Residual disease

Response categories - MRD

• MRD negative CR - Absence of aberrant clonal plasma cells by NGF or NGS on bone marrow aspirates with a minimum sensitivity of at least 1 in 10⁵ nucleated cells

• MRD rate at 10-6 rates should be reported when feasible designated as MRD negative CR 10-6

• If MRD negative as defined above but no negative immunofixation, should be defined as NGF/NGS negative (without CR) at either threshold

• Imaging negative MRD neg – MRD as defined above at either threshold with a functional imaging (PET-CT or WB-DWI MRI) negative

• Sustained MRD negative CR– MRD negative CR as defined above at either threshold with two negative MRD tests at least 24 months apart, and without any positive test in between

Conceptual – Deep or Stringent MRD

• MRD at 10-6 by NGS

• Negative by functional imaging (PET-CT or WB-DWI MRI)

• No monoclonal protein by mass spec

• No circulating plasma cells

• This will form the building block for a cure definition in future, when sustained for 5 years without therapy

Disease Progression

M protein measurements

• ≥25% increase in the level of the serum monoclonal paraprotein, which must also be an absolute increase of at least 5 g/l

• ≥25% increase in the difference between involved and uninvolved FLC levels. The absolute increase must be ≥10 mg/dl

• ≥25% increase in the 24 h urinary light chain excretion, which must also be an absolute increase of at least 200 mg/24 h

PD based on plasma cells/ plasmacytoma

•Definite increase in the size of existing bone lesions or soft tissue plasmacytomas (Deauville score> 4 (increase of SUV > liver) in (or MRI DWI RAC >= 3) in one or more existing lesion)

•≥25% increase in plasma cells in a bone marrow, which must also be an absolute increase of at least 10% only in patients without measurable disease by serum and urine

•Development of new bone lesions or soft tissue or para-skeletal plasmacytomas

•≥50% increase in circulating plasma cells (minimum of 200 cells per μL) if this is the only measure of disease

Additional points

• Confirmation of monoclonal protein measurements are required for every category, but sequential confirmation can be replaced by simultaneous confirmation – two measurements

• 24 hour urine measurements are required at baseline, and if M protein present, will need retesting only to confirm CR

Serum, Urine

More sensitive mass spectrometry (using expected protein sequence)

Marrow, Blood, Extramedullary

Circulating tumor/free DNA

Novel imaging techniques

Functional status

PANEL DISCUSSION

Panelists:

Elena Zamagni, MD, PhD

Bologna University &

Joshua Richter, MD

Tisch Cancer Institute, Mount Sinai

SESSION 3: NEWLY DIAGNOSED MYELOMA THERAPY

Session Chair: Philippe Moreau, MD

University Hospital Nantes

MINIMAL RESIDUAL DISEASE (MRD) ADAPTED THERAPY

Aurore Perrot, MD

University of Toulouse

MRD-adapted therapy

Aurore Perrot

University Hospital of Toulouse

Trial designs incorporating MRD-driven stratification

After induction

After transplant

Trial designs incorporating MRD-driven stratification

During maintenance

Perseus trial: MRD-based decision

Phase 3 Perseus trial

eligibility criteria:

Transplant-

Primary endpoint: PFSc

Key secondary endpoints: Overall CR rate,c overall MRD-negativity rate,d OS

Discontinue DARA therapy only after 24 months of D-R maintenance for patients with CR and 12 months of sustained MRD negativity

Restart DARA therapy upon confirmed loss of CR without PD or recurrence of MRD

Outcomes during maintenance

Maintenance

DARA: 1,800 mg SCb Q4W

R: 10 mg PO Days 1-28

MRD positive

Continue D-R until PD

MRD negative

Discontinue DARA therapy only

28-day cycles

Discontinue DARA therapy only

after 24 months of D-R maintenance for patients with CR and 12 months of sustained MRD negativity

Restart DARA therapy upon confirmed loss of CR without PD or recurrence of MRD

Phase 2 trial Master

Treatment interruption if « sustained » MRD negative

Phase 2 Master

Early interruption:

Not suitable for patients with high-risk cytogenetics

Phase 3 MRD-driven trial

Induction

MIDAS = MInimal residual Disease Adapted Strategy

MRD evaluation

x 6 1:1

IsaKRD x 6 (28d cycles) MRD

Stem cell collection after cycle 3 (G-CSF+/- plerixafor)

Risk-adapted consolidation and maintenance Standard risk (MRD <10-5) High risk (MRD >10-5)

ASCT + IsaKRD x 2

ASCT + IsaKRD x 2

(3 years)

Key eligibility criteria

NDMM < 66y

Transplant-eligible

ECOG 0-2

No active cardiac disease

. 791 patients included in 72 centers (Dec 2021 – Jul 2023) . 757 completed induction . 761 patients did at least one stem cell harvest

Randomized patient’s characteristics

High-risk

Perrot A et al, Blood 2025

Subgroup analysis

IsaKRD alone vs ASCT

(arm A vs arm B)

Subgroup analysis

Single vs tandem ASCT (arm C vs arm D)

Changes in MRD status (10

in arms A & B

Per protocol analysis

Changes in MRD status (10

, NGS), according to t(11;14)

Per protocol analysis

Aurore Perrot, MD

MRD kinetics according to CCND1 expression in Cassiopeia

Emerging Data from MRD-Driven Trials

MASTER trial: No early treatment discontinuation despite MRD negativity in patients with high-risk cytogenetics

MIDAS trial

– Can transplant be differed in MRD-negative patients after induction without PFS data?

– Tandem transplant can probably be abandonned

PERSEUS trial

Early relapses observed after discontinuation of daratumumab during maintenance?

 Open Questions

Should we consider different strategies for high-risk cytogenetics?

Could MRD after induction serve as an early stratification factor in slow responders such as patients with t(11;14)?

CURRENT APPROACHES TO NEWLY DIAGNOSED MYELOMA

Sagar Lonial, MD, FACP
Emory Winship Cancer Institute

Current Approach to NDMM

Emory University School of Medicine

Sagar Lonial, MD

Questions to Address Today

• What is the right regimen

• Is there a difference between transplant ineligible and frail?

• How long to treat

• Optimal endpoint and timing (early vs late)

• Are we ready to de escalate without clear cure?

Risk Stratification

• High risk

– Deletion 17p >20% and/or p53 mutation

– Deletion 1p and +1q (1 extra copy of 1q not high risk alone)

– High risk 14q32 trans and (+1q or deletion 1p)

• Standard risk

Hyperdiploidy

t(11;14) Is there an intermediate category?

Induction regimens in TE-patients with NDMM: Efficacy

1. Richardson PG, et al. New Engl J Med. 2022;387:132-147. 2. Voorhees PM, et al. Lancet Haematol. 2023;10(10):e825-837. 3. Sonneveld P, et al. N Engl J Med. 2024;3990(4):301-313.

Significantly Longer Estimated PFS Projections With DVRd vs VRd For the Best-fit Distribution

PFS (observed and projections)

DVRd-Exponential

VRd-Exponential

PERSEUS ITT

PERSEUS ITT

DVRd - PERSEUS ITT

VRd - PERSEUS ITT

DVRd-Exponential

VRd-Exponential

CEPHEUS TIE

CEPHEUS TIE

DVRd - CEPHEUS TIE

VRd - CEPHEUS TIE

Estimated PFS, DVRd vs VRd PERSEUS: 205 months (17.1 yrs) vs 87 months (7.3 yrs)

CEPHEUS: 100 months (8.3 yrs) vs 53 months (4.4 yrs)

Evolving standard of care for TIE patients with NDMM: Efficacy

MRD-negative

Median: 61.9 vs 34.4 months

60-month PFS rate: 52% vs 30% (P<0.001) 5-year rate: 63% vs 45% (P<0.001) 24-month rate: 85% vs 80% 54-month rate: 68% v. 50% (p<0.001)

De-escalation during maintenance

• Continuous therapy has carried the load

• Limited duration is the future

• Do we have the tools to define who has had enough therapy?

• Is the benefit of combinations for standard risk limited duration?

• follow up after discontinuation needs to be robust to detect failures

Is sustained MRD enough?

• If so how long and what interval and depth?

• Can all trials be reported in a similar way to enhance comparisons – Importance of fixed timepoints and consistent reporting

• Does MRD win over risk and genetics when considering duration?

Optimal endpoint

• Are Early and Late MRD the same?

• What is the role of PFS ?

• What is the role of OS?

• Are we ready to de escalate without knowing how many patients we are curing.

• As we seek to create more “patient friendly” regimens, how do we continue to improve outcomes (rather than staying the same?

Questions to Address Today

• What is the right regimen CD38 + IMID+PI

• Is there a difference between transplant ineligible and frail?

– Maybe but not as clear. More than age cut off

• How long to treat

– MRD directed may be of use, but follow up limited

• Optimal endpoint and timing (early vs late)

– Does escalation or de-escalation change natural history?

• Are we ready to de escalate without clear cure?

– Caution about losing the gains without long term follow up

Jonathan

Ajay

Nisha

Richa

Nishi

Craig

Vikas

Charise

Danielle

Donald

SESSION 4: UPDATE ON IMMUNOTHERAPY

Session Chair: Jesús San Miguel, MD, PhD

University of Navarra

OVERVIEW OF CURRENT STATUS OF APPROVED CAR T & BISPECIFICS

University of California San Francisco

CARS and Bispecifics: Current Status

Thomas G. Martin, MD

Clinical Professor of Medicine

Associate Chief, Division of Hematology

Co-Lead, Cancer Immunology and Immunotherapy

Helen Diller Family Comprehensive Cancer Center

University of California, San Francisco

San Francisco, California

Faculty Disclosure

Thomas G. Martin, MD

Clinical Professor of Medicine

Associate Chief, Hematology

Co-Lead, Cancer Immunology and Immunotherapy

Helen Diller Family Comprehensive Cancer Center

University of California, San Francisco Medical Center San Francisco, California

Thomas G. Martin, MD, has disclosed that he has received consulting fees from Pfizer, Lilly, AstraZeneca and GSK and funds for research support from Amgen, Johnson & Johnson – Janssen, Sanofi, and Seattle Genetics.

Amazing Success in Immunotherapy for MM

Munshi

Immunotherapy in MM: Going to Earlier LINES!!!!

Where will each agent have its BEST/Biggest Impact?

Treatment Attrition in Multiple Myeloma

• Real-world assessment of lines of therapy and outcomes based on 3 US insurance claims databases

Non-Transplant Patients

Will these drugs have curative potential in NDMM/Early RRMM!!!

A Mere 8 year ago………..

We need to interrogate the BM-ME

CARTITUDE PROGRAM

• CARTITUDE-1 (n=97) Accelerated FDA Approval: 2/22/2022. ?? Cure in RRMM

• CARTITUDE-2 (multi-cohort)

• A - Cilta-cel in patients with RRMM having received 1-3 PLT, Len refractory

• B – Cilta-cel in patients with RRMM s/p 1 PLT AND early relapse (<12m)

• C – Cilta-cel in TCE patients and exposure to BCMA therapy (non-CAR)

• D- Cilta-cel in NDMM patients without CR after Ind/SCT/Consol (4-8 cycles)

• E – HR-NDMM => D-RVd + Ciltacel + DR maintenance as frontline therapy

• CARTITUDE-4 (n=419) CART vs. SOC triplets Full FDA Approval

• CARTITUDE-5 (n=650) Phase 3: TI - VRD+Rd vs. VRD+Cilta-cel (Accrued)

• CARTITUDE-6 (n=750) Phase 3: TE – D-VRd/ASCT vs. D-VRD+ Cilta (Accrued)

Primary endpoint of PFS was met and study now reported 1. https://clinicaltrials.gov/ct2/show/NCT04181827 2.

https://www.prnewswire.com/news-releases/janssen-announces-unblinding-of-phase-3-cartitude-4-study-of-carvykti-cilta-cel-as-primary-endpoint-met-in-treatment-of-patie nts-with-relapsed-and-refractory-multiple-myeloma-301732398.html . San Miguel j. et al. NEJM 2023

CART-4: HR Subgroup Analysis

•Median follow-up 33.6 months

•Improved PFS in sub-groups

• LOT

• EMD

• HR cytogenetics

Study population

• 208 patients cilta-cel arm (ITT) and 176 received ciltacel (as-treated population)

• 211 patients SOC

CART-4: Results - Safety

Safety in the cilta-cel as-treated population

• CRS occurred in 76.1% of patients and were mostly grade 1/2; all cases resolved3,4 (Table 3)

• CAR-T cell neurotoxicity occurred in 20.5% of patients; none were fatal3,4

• ICANS occurred in 4.5% of patients; all were grade 1/2 and resolved3,4

• Cranial nerve palsy (9.1%), peripheral neuropathy (2.8%), and movement and neurocognitive treatment-emergent AEs (MNTs) (0.6%) were mostly grade 1/23,4

• By the CCO, all but 2 of the cranial nerve palsy and 2 of the peripheral neuropathy cases had resolved; the MNT case (grade 1) had not yet resolved by the CCO3,4

• Secondary Malignancies

Cilta-cel vs. SOC

Overall: 9(4.3) vs. 14 (6.7)

Cutaneous: 5 vs. 10

1st yr./ 2nd yr

3. San-Miguel J, et al. N Engl J Med 2023;389:335-47. 4. Palumbo A, et al. N Engl J Med 2016;375:754-66.

Teclistamab in 2024 ASCO+EHA

Phase1/2: single arm, Qwk => Q2/Q4wk 30-month mFu Toxicity

Elranatamab

Nooka Et al.

Elranatamab After Previous BCMA Therapy

• Pooled analysis of 4 MagnetisMM trials; median time to response 1.7 mo (range: 0.3-9.3) • Response

Bispecific Antibodies With Other Targets

GPRC5D Bispecifics: single agent and in combination

Bispecific Antibody

Anemia

Thrombocytopenia

ICANS #

Hypogamma/IVIg Other

(0.7%)

(0%)

(2.0%)

(28%)

(53%)

(25%)

(29%)

(1.8%)

0 due to AEs NR/10%

Dysgeusia 77% (N/A)

(0%)

63% (0%)

(0%)

(26%)

(26%)

(20%)

(0%) 1 due to AE

(4%)

(2%)

(3.2%)

(53%)

(61%)

(34%)

(29%)

(? 1)

(N/A)

(0%)

(0%)

Methods: LINKER-MM2 (NCT05137054) study design : Linvo + K(56) - RRMM

• Phase 1b trial conducted across 39 sites in USA, France, Greece, and Spain

Inclusion criteria:

• Age ≥18 years

• RRMM

• ≥3 LoT, or ≥2 LoT if either TCE or DCR (IMiD + PI)

• Prior CFZ treatment permitted if previously tolerated and ≥6 months had elapsed since last exposure

• CFZ-refractory patients were allowed during dose finding

• DLTs • Incidence/severity of TEAEs

Belantamab

ASCO 2025 Mateos et al.

HR Cyto = Median PFS 33.2 mo (95% CI. 20.1-NR)

Median OS NR (18 mo CI. 80%)

Immunotherapy Bispecific Trials

Current and planned (not inclusive of all trials)

MajesTEC-7: Tec-D vs. DRd

Myeloma Treatment Paradigm

M-Tec-3: Tec-D vs. DPd/DVd M-Tal-1: Tal SQ

Mag-6: Elran-DR vs. DRd M-Tec-4: R vs. Tec vs. Tec-R

Linvo: Phase I/II

Frontline – maintenance. Early RR RRMM (TCE)

Mag-7: Elran vs. Len

Linvo: Linker MM-4

Linvo: Combinations

Mag-3: Elran (single)

Mag-5: Elran, Elran +D, Dara+Pd

Camma-1: Cevo, CevoPd, CevoDd

Biomarkers and resistance important for all timepoints

Camma-3: Cevo SQ

Frontline: time-limited, no CART!

Quach, H et

Results: MagnetisMM-6

Frontline: Which Novel IT, when, how long?

Induction

Len +/- CD38

Maintenance

BiSp +/-CD38

Maintenance

BiSp +/-CD38

Maintenance

Len +/- CD38

Maintenance QUAD Induction

QUAD -BiSp

Triplet/Quad Induction

BsAb +/- partner

BiSp +/-CD38

Maintenance

BsAb (different target) Bela maintenance

QUAD -Bela Bela +/- partnermaintenance

Conclusions:

• CAR is currently the front-runner for BCMA Therapy for RRMM in BCMA Naïve patients

• Is there room for more CARs  YES

• Wait for Hermann’s talk!!!

• Convenience and lowest TRM/morbidity and best QOL

Frontline is the space with the greatest potential impact

• “Cures” win – whatever the platform/Sequence

Discussion  about the FDA:

• We shouldn’t change our practices/advances based on the changes at FDA...

• We should continue our partnership and collaboration

• We need to solidify endpoints  OS, PFS, MRD

• Can we agree on a definition of “CURE” ?

Discussion  about the FDA:

• We shouldn’t change our practices/advances based on the changes at FDA...

• We should continue our partnership and collaboration

• We need to solidify endpoints  OS, PFS, MRD

• Can we agree on a definition of “CURE” ?

• Example from last Thursday 6/5/2025

• IND 31367

• Submitted 5/5

• Approved 6/5

Adapting the TRAC concept to Myeloma

And optimize the CAR design

Commercial CARs

CARVYKTI

TRAC-CARs

Clinical scale manufacturing UCCAR-1

1XX based on Feucht et al., Nat. Med 2019

Talbot et al., unpublished. Shy et al. unpublished

SEQUENCING IMMUNOTHERAPIES

Nizar Bahlis, MD

University of Calgary

Sequencing immunotherapies in myeloma

University of Calgary

IMWG 2025 Summit, Milan

Both TCE and CAR T induce proximity-mediated formation of immunological synapse

Cytolytic synapses induced by BsAbs or CART have the same composition, subdomain arrangement and size from synapses induced by regular components of cytotoxic T cell recognition.

Cytolytic activity of BsAb is largely mediated by granzymes

granzymes as well as Death Receptor ligands

Pourbrahim M. unpublished data Boise L provided Serpin B9 and FADD-KO cells

BsAb

(CD3

x Ag)

activate

effector memory T cells (TEM) but not naïve T cells

CAR T cells equally activate naïve and memory T cells

Scheller L et al, Leuk Lymphoma. 2024 Feb

in the drug

product correlated with longer PFS in patients treated with Ciltacel

Bispecific vs CAR T: T cells effect and dependencies

Efficacy

Bispecific CAR T

Activate Tem >>> T naïve

+++ CD8 >> CD4 dependent

↑ absolute CD3 count

↓ Texhaused (TCF1 - TOX +)

Higher E:T ratio +++

Higher CD3/sBCMA ratio +++

Efficacy reduced with ↑ Tregs

Equally activate Tnaive & Tem

↑ Tnaive/Tcm in leukopheresis ++

↑ Tem in product +++

CD4/CD8 ratio 50/50 leukopheresis and product

Higher E:T ratio +++

Higher Cmax/sBCMA ratio +++

Efficacy reduced with ↑ Tregs

Effect on T cells

CD8 Tem /TEMRA ↑

CD4 / CD8 ratio ↓

Lymphodepletion (BM pooling)

diversity ↓ CD4 / CD8 ratio ↓ (LD chemo) Lymphodepletion (LD chemo)

diversity ↓

FACT #1: TCE treatment reduces CD4/CD8 ratio and modifies TCRs repertoires (lower

diversity)

FACT #2

Significant overlap between T cells activation and exhaustion phenotypic and transcriptomic markers

Patient P3796

need to demonstrate functional exhaustion

Healthy donor (HD) or patient (SW) peripheral blood mononuclear cells PBMC) co-cultured with OPM2 cells (CTV stained) with or without elranatamab (ELRA) or alnuctamab (ALN). Effector to target ratio 10:1. OPM2 viability was assesses at 48 hours.

E:T ratio and absolute lymphocytes count highly impact the efficacy of TCEs

FACT #4 Target antigen loss is the major mechanism of acquired resistance to anti-BCMA/ –GPRC5D TCEs and anti-GPRC5D CAR T

(NOT the case post anti-BCMA CAR T)

Very frequent and convergent evolutional patterns of TNFRSF17

subclones post anti-BCMA BsAb

A.BCMA antigen escape = 24 / 40 (60 %)

GPRC5D antigen escape is identified in 61.9% of cases relapsing after talquetamab (13/21)

Lee H,…Bahlis N et al. Manuscript

Sequencing immunotherapies in MM

No target Ag switch:

1. Anti-BCMA TCE / ADC  anti-BCMA CAR

2. Anti-BCMA CAR T / ADC  anti-BCMA TCE

Target Ag switch:

3. Anti-BCMA TCE  anti-GPRC5D TCE or CAR T

4. Anti-BCMA CAR T / ADC anti-GPRC5D TCE

Anti-BCMA TCE or ADC  anti-BCMA

Patient 1:

65 yo female Multiple Myeloma IgG kappa R-ISS III

Treatments:

1.Dara-VTD  Mel 200 VGPR; DOR ~ 14 months

2. Carfilzomib_Len-Dex VGPR; DOR ~ 23 months

3. Dara_Pom_Dex  VGPR; DOR ~ 21 months.

4. Teclistamab + Cetrelimab ~ 20 months remission

71 yo with PENTA REFRACTORY MM including anti-BCMA TCE

5. Ide-Cel ~ 3 months post anti-BCMA TCE, March 26, 2024

ongoing remission ~ 15 months

Convergent evolution with TNFRSF17 mutant clones

WGS at progression post Teclistamab :  3 TNFRSF17 mutant clones:

• Clone #1: TNFRSF17 p.R27Q (chr16:11965404) VAF 20.51%

• Clone #2: TNFRSF17 p.C24Y (chr16:11965395) VAF 13 %

• Clone #3: TNFRSF17 c.del 86-90 fs del with early stop codon VAF 10%

TNFRSF17 monoallelic deletion chr16:11825306-12262140

(size = 436.83 Kb ; log2 = -0.93)

Treatments:

1.Dara-VTD  Mel 200 VGPR; DOR ~ 14 months

2. Carfilzomib_Len-Dex VGPR; DOR ~ 23 months

3. Dara_Pom_Dex  VGPR; DOR ~ 21 months.

4. Teclistamab + Cetrelimab (PD1 inhibitor) ~ 20 months remission 5.

Anti-BCMA TCE  anti-BCMA

CAR T recommendations

 Allow Sufficient washout time for lymphocytes count recovery and adequate CD4/CD8 ratio prior to apheresis; may vary between patients (role for IMiDs , CELMoDs, stem cell boost?)  optimal CAR-T product

 Confirm retained target expression (FCM and/or NGS)

 Bridging with TCE prior to CAR T infusion does not negatively impact outcomes (Fandrei D, Blood Cancer Discovery 2024, Dhakal B et al Blood 2025))

Sequencing immunotherapies in MM

No target Ag switch:

1. Anti-BCMA TCE / ADC  anti-BCMA CAR

2. Anti-BCMA CAR T / ADC  anti-BCMA TCE

Target Ag switch:

3. Anti-BCMA TCE  anti-GPRC5D TCE or CAR T

4. Anti-BCMA CAR T / ADC anti-GPRC5D TCE

Teclistamab in

patients with relapsed/refractory multiple myeloma after BCMA-targeting therapies

mDOR 14.8 mos (95% CI , 6.222.6)

mPFS 7.3 mos (95% CI, 1.316) Touzeau C et al Blood Aug 2024

PD-1 & TIM-3 were NOT upregulated on CD4+/CD8+ T cells from patients receiving a prior anti-BCMA CAR-T or ADC

Touzeau C et al Blood Aug 2024

RWA teclistamab in RRMM patients in German cohort:

Patient 2:

65 yo male Multiple Myeloma kappa light chain R-ISS II, t(11;14)

positive Treatments:

1.CyborD  Vel- Mel 200 Feb 2015 Len maint 3 yrs VGPR; DOR ~ 4 years

2. Dara_Len_Dex , Feb 2019  VGPR, PFS ~ 1 year

3. BB2121-MM03 trial, Ide-Cel, Aug 2020, cell dose 563 x 10^6. SD PD Dec 2020

4.TECLI-MMY1001 trial with Teclistamab Jan 20, 2021. Best response as sCR ~ 1 year. PD June, 2022

5.TRIMM-2: Daratumumab +Talquetamab + dexamethasone, pomalidomide. Sep 70 yo with double refractory MM progressed 3 months post antiBCMA CAR

p.Ser30del

Lee H et al. Nat Med. 2023;29:2295-2306

Sequencing immunotherapies in MM

No target Ag switch:

1. Anti-BCMA TCE / ADC  anti-BCMA CAR

2. Anti-BCMA CAR T / ADC  anti-BCMA TCE

Target Ag switch:

3. Anti-BCMA TCE  anti-GPRC5D TCE or CAR T

4. Anti-BCMA CAR T / ADC anti-GPRC5D TCE

Bispecific antibodies targeting BCMA or GPRC5D are highly effective in relapsed myeloma after CAR T-cell therapy

MonumenTAL-1: PFS in patients with TCR

Patient 2: 65 yo male Multiple Myeloma kappa light chain R-ISS II, t(11;14) positive Treatments:

1.CyborD  Vel- Mel 200 Feb 2015 Len maint 3 yrs VGPR; DOR ~ 4 years

2. Dara_Len_Dex , Feb 2019  VGPR, PFS ~ 1 year

3. BB2121-MM03 trial, Ide-Cel, Aug 2020, cell dose 563 x 10^6. SD PD Dec 2020 4.TECLI-MMY1001 trial with Teclistamab Jan 20, 2021. Best response as sCR ~ 1 year. PD June, 2022

2022. sCR ongoing ~ 4 years 70 yo with double refractory MM progressed 3 months post anti-

Treatments:

1.CyBorD

2.Dara_Len-Dex

Double hit, penta refractory MM

Therapy: anti BCMAxCD3

IMWG response: CR; DOR 13 months

Elranatamab

Lee H et al. Nature Medicine. 2023;29:2295-2306

TNFRSF17 mono-allelic loss with extracellular domain mutation

Lee H et al. Nature Medicine. 2023;29:2295-2306

Differential sensitivity of BCMA mutant clones to BCMA xCD3 TCE

75 yo female Multiple Myeloma IgG Lambda R-ISS III

t(14;16) pos

Treatments:

1.CyBorD VGPR; DOR ~ 18 months

2.Dara_Len-Dex VGPR; DOR ~ 16 months

3.Pom_Dex  MR; DOR ~ 4 months.

4.Carfilzomib_Dexamethasone  PR; DOR ~ 14 months + del17p 50.8%

80 yo with Double hit PENTA REFRACTORY MM

5. Elranatamab 76 mg  sCR; DOR ~ 13 months

6. Talquetamab q2w, Daratumumab & pomalidomide (2 mg added C2D1)

sCR; , DOR ~ 13 months

Anti-HA Anti-GPRC5D

C. Dose response cytotoxicity assay

GPRC5D D239 forms ionic interaction with CDR3 of talquetamab heavy chain

Shallow pocket in GPRC5D extracellular domain comprised of: - ECL 1, ECL2, TM3, TM5, and TM7 - Walls of the pocket formed by:-

R102 of talquetamab intrudes into pocket forming an ion pair with D239 of GPRC5D

Anti-BCMA CART or ADC

or anti-GPRC5D TCE

 Confirm retained target expression (FCM and/or NGS)

 Ensure adequate absolute CD3 T cell count (CD8 Tem)

 Sequential treatment with anti-BCMA CAR –T  TCE or TCE

 TCE can yield durable responses if adequate T cell count (Tem)

 Switching targeted antigen with Talquetamab if TNFRSF17 mutational status is unknown.

Additional questions to be answered

 Optimal duration of therapy with bispecific antibodies

 Better define patients who respond to sequential TCE based therapies?

 Immune profiling

 Should we combine TCEs targeting different MM Ag? Combine TCE +/- anti-CD38 mAb to target antigenic escape? Or with CelMODs to expand Tem?

 Surveillance for emerging BCMA or GPRC5D mutants

switching treatment

NEXT

GENERATION CAR T, BISPECIFICS & TRISPECIFICS

FRCP

Würzburg University Hospital

June 10th 2025, Milan

Next Generation CAR-T, Bispecifics and Trispecifics

Prof. Dr. Hermann Einsele Department of Internal Medicine II University Hospital Würzburg

Why do we need novel constructs and novel strategies for T Cell engaging therapies?

To improve efficacy

• T-cell exhaustion

• Target antigen loss

• Tumor microenvironment

To reduce Toxicities

CRS

ICANS

CNP/MNT

To improve availability

Long manufacturing time (CAR-T Cells)

Cost

To improve Efficacy /

Safety

Median (95% CI): 8.8 mo. (5.6 – 11.6)

CRS grade >2 3-5%

ICANS grade >2 3-5%, CNP/MNTs 1-10%

Bispecifics:

Require hospitalization for step-up dosing

Infection Rate Grade >2 40-70% Safety Majestic-1

Why do we need novel constructs and novel strategies

For T cell engaging therapies?

To improve efficacy

• T Cell exhaustion

• Target antigen loss

• Tumor microenvironment

To reduce Toxicities

CRS

ICANS

CNP/MNT

Combinations

Novel constructs

(binders, modulation of Transcription factors, Inhibitory molecules)

To improve availability

Long manufacturing time (CAR-T Cells) • Cost

Combination with small molecules, e.g. Dasatinib

(BTKi,

Novel Constructs: Anitocabtagene Autoleucel BCMA CAR-T cells

High Efficacy – Low Toxicity

Small D-Domain construct facilitates high transduction efficiency and CAR positivity, with low total cell dose

D-Domain CARs stable and lack tonic signaling

D-Domain binder fast off-rate, promoting tumor cell killing without prolonged inflammation

→ reduced neurotoxicity?!

At median follow-up of 34 months:

ORR 100%, CR 79%

Median PFS for all pts. 30.2 mo. / for CR/sCR pts. 34.3 mo.

No delayed or non-ICANS neurotoxicities

(Parkinsonism, cranial nerve palsies, GBS)

CRS 95 % (≥ Grade 2 47%), ICANS 18% (Grade ≥ 2 6%)

Re-wiring CAR-Ts through modulation of transcription factors

To install genetic program for sustained anti-myeloma efficacy and longevity in CAR-Ts,

Rejuvenate CAR-Ts

• restoration of IL2-production

• exhaustion resistance ↑

• terminal differentiation ↓

• potency in Aglow settings ↑

Knock-in screening

Batf3-deficiency

Memory formation (transition-to and quality-of) ↓

contraction after response ↑ pro-apoptotic factor BIM ↑

100 TFs (and TFxTF) in TCR and CAR + different stimulations  TFAP4 (+BATF/BATF3) promotes proliferative, stem celllike memory state

Overexpression of relevant Transcription Factors to enhance

T Cell stamina and memory

Transcription factor Batf3 regulates T-cell apoptosis and longevity via the proapoptotic factor BIM  Batf3-transduced CAR-T Cells targeting ROR2 in r/rMM

Single Epitope Targeting Immunotherapies

BCMA Antigen Escape

BCMA-CART Cells

BCMA-targeting TCE

Whole genome sequencing

4-6 % irreversible BCMA loss Biallelic deletion TFRSF17

GPRC5D-targeting T cell engaging therapies

After BCMA -T cell therapy

Arlo-Cel for r/rMM (No/prior BCMA)

Response in TCR-naïve and -exposed cohorts1

0.8 mg/kg Q2W cohort, 23.4-month mFU; Prior TCR cohort, 20.5-month mFU ORR: 66.7% (n=52/78)

Bal S. et al., ASH 2024

Chari A. et al., Lancet Haematol 2025

Talquetamab for r/rMM (no/prior BCMA) Lee et al.,

Cevostamab for r/rMM - FCRH5 targeting (no/prior BCMA)

Library. Kumar S. 06/13/2024

But: GPRC5D-targeting T cell engaging therapy is also associated with target antigen loss. 13/17 cases relapsing during Talquetamab therapy and 8/10 patients relapsing after Arlo-cel showed GPRC5D Ag Escape

EHA

Novel Targets: CAR T Cells

CARAMBA-1: SLAMF7-directed CAR T Cells

SLAMF7 is a strong CAR-T target in MM

• Sustained high level expression on MM/EMD,

• no interference from soluble SLAMF7

CARAMBA-1 is a First-in-Human Phase I/IIa trial of SLAMF7 CAR-T therapy Dose escalation is ongoing

• Safety: favorable safety signal, no DLTs

• Efficacy: SLAMF7 CAR-T engraftment, responses in heavily pretreated MM

• But: SLAMF7 Expression on activated T Cells / CAR-T Cell fratricide

Two additional Arguments for SLAMF7 CAR-T cells

SLAMF7 on … bone marrow fibroblasts & macrophages (1)

SLAMF7 on … NKT cell lymphoma & T cell lymphoma after CAR T Therapy(2)

Modified from Sakemura, Blood 2022

Elmagaacli et al. Leuk Lymphoma 2019

A Novel Target in Multiple Myeloma: ROR2

ROR2 expression early stages of embryogenesis

Absent on most adult-human tissues

Oncofetal antigen – associated with invasiveness

90 % of primary MM samples show ROR2 expression above threshold

ROR2-specific CAR-T cells elicit long-term tumor control and a curative effect on mice treated in a model of advanced MM (U-266 xenograft)

J. Weber, M. Hudecek; unpublished data; do NOT post

Dual Targeting CAR-T Cells to reduce the risk of Target Ag loss

Dual-targeting BCMA-CD19 (Gracell): FasTCAR-T GC012F

BCMA/GPRC5D bispecific CAR-T Cells

Bispecific Antibodies

Dual targeting with ISB 2001 (BCMAxCD38xCD3):

First TREATTM Trispecific Antibody for r/rMM

Deep and Lasting Responses Observed at ≥ 50 μg/kg /

Dose Escalation continues

The Next Generation of Targeted Immunotherapies reduces Side Effects and Enhances Efficacy in r/rMM (4

Novel Bi-Molecular T Cell Activating Antibodies

Antibody mediated therapy today

Hemibody Solution

• IgG

• BiTEs, DARTS, Tribodies etc.

Problem

• No singular tumor antigens

• Poor specificity

• Active CD3 binding side

• Unspecific T Cell activation

• CRS /ICANS

• Higher specificity

• Targeting a broad range of Ags

• No unspecific T Cell activation

• Prevent CRS /ICANS)

• Reduce Infections

T Cell engagement induced by Hemibodies and BiTEs

Banaszek, A. et al., Nat Comm 2019 Geis, M., et al., Commun Biol 4, 44, 2021

CHY001: Hemibodies co-targeting SLAMF7/CD38 for Multiple Myeloma

CD38 and SLAMF7 are stably expressed on MM

• Dual CD38/SLAMF7 expr. exclusively on MM cells

• All CD138 pos. MM Cells strongly express CD38 and SLAMF7

• No co-expression on T, NK or hematopoieticprogenitor cells

Patients >18y suffering refractory/relapsed (RR) MM, n=30

• No Ag shedding, very low risk of target antigen loss

Inclusion criteria:

• Measurable disease after 3rd line therapy

• Allowed: proteasome Inhib., Imids, ASC, including CD38 and SLAMF7 antibodies

• Prior CAR-T or bispecifics allowed except for anti CD38 or SLAMF7 directed T Cell engaging therapies

Primary endpoint:

• Determination of safety and tolerability (AA/SAA according to CTCAE V5.0)

• MTD and DLT

Secondary endpoints:

• Efficacy: ORR/ CR-rate/MRD negativity

Focal Infiltration of Immune Cells in EMD Lesions

(Spatial Transcriptomics)

• EMD lesions are infiltrated by immune cells that are anatomically confined to distinct niches.

• T Cells found in the vicinity of PCs showed signs of T Cell dysfunction.

John M., Rasche L. et al., Blood.

Next Generation Genetic Engineering:

Base/Prime Editing TRUCKS, the fourth-generation CAR-T Cells

Transgenic proteins released by chimeric antigen receptor (CAR) T Cells upon activation impact tumor environment

H. et al., Adv Cell Gene Ther. 2020

Abken,

Enhanced CAR-T Cell Therapy for Lymphoma

after Previous Failure Improving expansion and persistence

Three-Month Response (all patients and by lymphoma subtype)

Responses in patients with NHL resistant to CD19-CART cell therapy

huCART19-IL18 Expansion and Persistence According to Dose Level

Shorter manufacturing time Humanized CD19 CAR Local IL-18 production

3/4 patients evaluable showed CAR-T Cell persistence > 2 yrs

Median DOR: 9.6 mo.

Additional Challenges for CAR-T Cell Therapy

• Manufacturing time for CAR-T Cells: 6-8 weeks

 10-15 % do not receive CAR-T (progression/death while awaiting manufacturing)

• High Costs  restrictive use of CAR-T Cells

Strategies:

• More rapid Generation of autologous CAR-T Cells

• Academic CAR-T Cell production (ARI002h, SLAMF7)

• Allogeneic CAR-T Cells

• In vivo CAR-T Cell production

PHE885: Rapid Production and Expansion in Vivo

MRD Negativity Rate

T-charge

• Innovative platform for CAR-T

• Reduces manufacturing time < 2d

• Fully human BCMA CAR

• Preserves T Cell stemness

• Long presistence (71% at 12 mo.)

• Deep responses 100% ORR (up to 100% MRD neg,)

• Ongoing for > 2 years in several patients

Allogeneic CAR-T Cells

= off the shelf CAR-T Cells

Extensive genetic engineering

• Function?

• Persistence?

Allogeneic cells

• Immune-mediated rejection

• Persistence?

Cells = Off-the-Shelf CAR-T Cells

In the FCA 320M CAR+ Cell dose group, 17 patients (71%) ORR

11 (46%) VGPR+

6 (25%) CR/sCR Median Duration of Response: 8.3 mo.

In Vivo Gene Immunotherapy (in Vivo)

Mai, D. et al., Sci. Transl. Med. 14, eabo3603 (2022)

Main vector systems employed for in vivo CAR-T generation

Viral-based

non-viral

mRNAbased

From: Bui et al. (2024) eBioMedicine 106, 105266

novel transposon-LNPs

mRNA & mcDNA co-encapsulated

Many challenges are associated with main in vivo CAR-T technologies

General challenges of in vivo CAR-T approach Challenges with viral vectors Challenges with mRNA-LNPs

Precise targeting of T cells

General Safety due to off-target gene delivery

Distribution to / loss of vector in highly vascularized organs

Need for GLP-tox studies in relevant tox species (NHPs ?)

Efficiency of CAR-T generation, are sufficient numbers possible ?

Immunogenicity

LV tropism for other cells (esp. hepatocytes)

Anti-viral immunity (incl. pre-existing for certain AAV serotypes)

LV & RV: generally secondary leukemias

Repeat dosing required due to transient expression of mRNA

toxicity of LNP components (PEG, cationic lipids)

Repeat dosing causes increased immunogenicity

Possibility of generating fatal CAR-leukemia cells efficiency of gene transfer into resting T cells

LV/RVs need proliferating cells for efficient vector integration

Viral vectors are still complex to manufacture and expensive

Transposon-LNP technology combines the «best of both worlds»: (i) stable CAR expression ( LVs), (ii) scalable CAR-Ts ( mRNA-LNPs), but safe

In vivo generation and immunotherapy of CAR-T cells

Process of in-situ CART-T therapy with nanoparticles in vivo

+ Transposase (Sleeping Beauty)

Gene transfer into resting T cells + local Cytokine production

Next Generation CAR-T and T Cell engaging Antibodies

• Combination therapies (TKIs, IMiDs, CELMoDs, ICPi etc.) will further improve persistence and the efficacy of CAR-T Cells/ T Cell engaging Antibodies

• Additional MM targets and multitargeting to reduce antigen loss (Novel targets, Dual CAR-T, trispecific Antibodies)

• More rapid and less expensive production of CAR-T Cells (Academic CAR-T production, allo CAR-T)

• In Vivo generation of CAR-T Cells to increase global availability (off-the-shelf, reduced cost)

Thanks to

Medizinische Klink/Poliklinik II, Würzburg

M. Topp M. Hudecek

T. Bumm S. Danhof

L. Rasche M. Kortüm

G. Stuhler A. Beilhack

J. Waldschmidt T. Steinbrunn

Immunologie, Würzburg

W. Kastenmüller G. Gasteiger

Medizinische Klinik III, Regensburg

W. Herr M. Edinger

Med. Mikrobiologie, TUM

D. Busch

Universitätsklinikum Heidelberg

M. Raab H. Goldschmidt

E. Mai

UKE Hamburg

K. Weisel L. Leypoldt

Wilhelminen Cancer Research Institute, Wien

H. Ludwig

Zelltherapie u. Immunologie, Leipzig

U. Köhl

Medizinische Universität Wien

M. Krauth

Princess Margaret Cancer Centre, Toronto

K. Stewart S. Trudel

University of Calgary

N. Bahlis P. Neri

H. Lee

Università di Torino

F. Gay M. Boccadoro

R. Mina

Ospedale San Raffaele, Mailand

M. Casucci C. Bonini

Memorial Sloan-Kettering Cancer Center, NYC

S. Usmani F. Maura

M. Merz

Dana-Farber Cancer Institute, Boston

N. Munshi K. Anderson

S. Treon C. Mitsiades

Fred Hutchinson Cancer Research Center, Seattle

S.R. Riddell M. Jenssen

UCSF Helen Diller Family Comprehensive Cancer Center

Tom Martin A. Chari

Myeloma Research Rotterdam

P. Sonneveld T. Cupedo

A. Broijl

Amsterdam UMC

N. van de Donk S. Zweegman

Patients and family members

Funding Agencies

Many thanks for your kind attention!

University of California San Francisco

Mechanisms of Resistance to Immunotherapy

Professor of Clinical Medicine

Director of Multiple Myeloma Program

Co-Director of Clinical Research Hematology/Oncology

Helen Diller Comprehensive Cancer Center

University of California, San Francisco San Francisco, California

Disclosures

Research Support/P.I. Janssen Employee N/A

Consultant/Scientific Advisory Board

Abbvie, Adaptive, Amgen, Antengene, Bristol Myers Squibb, Forus, Genetech/Roche, Glaxo Smith Klein, Janssen, Karyopharm, Millenium/Takeda,

Sanofi/Genzyme

Tumor Intrinsic

T cell Health

Treatment Characteristics

Microenvironment

Overall Response Rate (ORR) and Progression Free Survival (PFS) of Recently Approved Therapies in RRMM

T cell redirection therapies

For > 4 LOT and IMID/PI/anti CD38 exposed

Siegel DS et al. Blood 2012;120(14): 2817–2825

Lonial S et al. Lancet 2016;387:1551-1560

Chari A et al. N Eng J Med 2019;381:727-738

Rasche et al EHA 2024

Van De Donk et al IMS 2023

Lesohkin et al Nat Med 2023

Anderson L et al. ASCO 2021;abstract 8016 (poster presentation)

Usmani S et al ASCO 2022;abstract 8028 (poster presentation)

Overall

Response Rate (ORR) and Progression Free Survival (PFS) of Recently Approved Therapies in RRMM

For > 4 LOT and IMID/PI/anti CD38 exposed

Lonial S et al. Lancet 2016;387:1551-1560

Chari A et al. N Eng J

Rasche et al EHA 2024

Van De Donk et al IMS 2023

Lesohkin et al Nat Med 2023

Anderson L et al. ASCO 2021;abstract 8016 (poster presentation)

Usmani S et al ASCO 2022;abstract 8028 (poster presentation)

Tumor Intrinsic Resistance: BCMA Extracellular Domain Antigenic Alterations

Impact of BCMA-ADC on subsequent Anti BCMA therapies

1. Anderson L et al. 2021 ASCO. Abstract 8016. 2. Ferreri CJ et al. 2021 ASH. Abstract 766. 3. Berdeja J et al. Lancet. 2021;398;314-324. 4. Lin Y et al. 2022 EHA. Abstract P961. 5. Cohen AD, et al. Blood Adv. 2019;3:2487-2490;

• longitudinal monitoring of immune cell populations showed no change in immune cell ratios or major cell populations, regardless of response status

• sBCMA levels showed pronounced drop during response but returned to baseline upon progression

• Impact on functional loss of BCMA unclear

Tumor Intrinsic Resistance: GPRC5d alterations Post Talquetamab

• In NDMM (COMMPASS) Monoallelic loss of GPRC5d on chromosome 12p: 13.17%

• 8/9 patients had GPRC loss after treatment with talquetamab

• 2 had biallelic loss

• 6 had SNV +/- monoallelic loss

• Many GPRC5d mutations result in ER trapping and loss of membrane trafficking

• 1 had alteration in ionic binding with GRPC5d and talquetamab

• Epigenetic silencing also possible

T cell Health Related Resistance:

Teclistamab Outcomes by Lines of Therapy

• Efficacy/Primary refractoriness appears to improve in earlier lines of therapy

• As compared to NDMM, RRMM have lower CD4/CD8 ratio and CD8 + clonality (Friedrich et al Cancer Cell 2023)

MRD Negative vs. MRD positive post CART

CARTITUDE 2: at apheresis, majority CD4+ T cells central memory; CD8+ T cells CM & stem-cell like memory and TEMRA cells - T cell phenotypes similar in pts with prior ADCs vs prior BsAB

T cell Health Related Resistance: Anti-BCMA Therapies Prior to GPRC and FcRH5 Bispecifics, Target Switch Not Enough

Talquetamab Phase ½

MonumenTAL-1 Study

(phase 1 study)

(Phase ½

)

• At baseline, prior TCR pts have less favorable immune phenotype vs TCR-naïve

• prior CAR-T pts have greater T-cell activation in 1st cycle vs BsAb group

• Preliminary data shows marked improvement with Dara + talquetamab PFS 19 mos (including prior bsAB resistant pts)

T cell Health Related Resistance: Inhibitory Immune

Checkpoint Expression by Blood phenotyping

 In the prior BsAb group, a higher PD1+ CD8 % cells across all T-cell subsets, and a higher frequency of TIGIT+ CD8 T cells for TSCM and Tnaive, suggesting a systemic T-cell dysfunction

 12-week washout required between discontinuation of last prior BsAb therapy and 1st cevostamab dose in CAMMA 2 may not be sufficient to overcome this dysfunction

% PD1+ CD8+ T-Cell Subsets

% TIGIT+ CD8+ T-Cell Subsets

* *

Prior ADC (n = 7) Prior CAR-T (n = 9) Prior BsAb (n = 17)

Microenvironment Resistance: High Inflammatory State & Cytopenias Correlate with Worse Efficacy

CART-hematotox score: >2 high risk (Hthigh) vs 0–1 low risk (HTlow)

One point : ANC ≤ 1.2, Hg ≤ 9, plt 76–175k, CRP ≥ 3.0 mg/dl, ferritin 650 to 2000 ng/ml.

Two points: plt ≤ 75 ferritin ≥ 2000 ng/ml.

•CAR-Hematotox higher scores correlate with worse efficacy:

•inferior ORR 44% vs. 70% p = 0.01)

•inferior PFS (median 5 vs. 15 mos, p < 0.001)

•inferior OS (median 10.5 mos vs. NR, p < 0.001)

Rajeski et al. J Hematol Oncol . 2023 Jul 31;16:88

Microenvironment Restistant: MM patients with shorter PFS Post CAR-T have significant increase in MDSCs

Analysis of patients receiving BCMA directed CAR-T therapy

• S100A8 is one of most elevated transcripts in MDSCs in MM pts

• S100A8 is an alarmin, part of the DAMP (Damage

Associated Molecular Pattern) family released in response to tissue injury

• Secreted by myeloid cells, mesenchymal cells, PMNS

Microenvironment Resistance: Blood S100A8/A9

levels increase at Month 1 in rapid progressors

Microenvironment Resistance: Blood S100A8/A9

levels increase at Month 1 in rapid progressors

• the decrease of CAR-T cytotoxicity with addition of S100A8/A9 is increased in a dose dependent manner

• suboptimal responders to bispecifics also have higher S100 at baseline and during therapy

• S100 promotes T reg differentiation

• addition of anti-S100A8/A9 mAb erases S100

inhibitory impact in both CART & bispecifics and also reduces T cell exhaustion

S100+ Myeloid cells are surrounded by TOX+ T cells in Extramedullary Myeloma after TCE therapy

CD3+ TOX negative T cells

CD3+ TOX+ T cells

CD14+ S100A8/A9 negative cells

CD14+ S100A8+ cells

CD14+ S100A9+ cells

T cell Engagers

Alnuctumab (BCMA) Forimtamig (GPRC) Abbv-383 (BCMA) EMB06(BCMA) JNJ-79635322 (BCMA* GPRC)

extender 2:1 tumor:CD3 binding domain low CD3 affinity trispecific tetravalent

Treatment Characteristics of Resistance: Bispecific and Trispecific Engagers

BCMA CAR-T cells in Multi-Drug Refractory MM

PFS 30 mos

Phase 1 (n=38)

1. Anderson L et al. ASCO 2021;abstract;8016 (poster presentation); 2. Berdeja J et al. Lancet 2021;398;314-24; 3. Lin Y et al. EHA 2022;abstract P961 (poster presentation);4 Freeman C et al ASH 2024. 5. Costello C, et al. ASH 2020;abstract 134; 6. Mohyuddin GR et al. Blood Adv 2021;5(4):1097-1101; 7. Oliver-Caldés A et al. ASH 2023

8. Li C et al. EHA 2022;abstract S187 (oral presentation); 9. Li C, et al. ASCO 2023; 8025 10. Mailankody S, et al. ASH 2021;abstract 651; 11. Dohlaria et al ASH 2023; abstract 3479.

CAR-Ts In Development

Novel manufacturing

• Dd domain, piggy Bac, human scFV, allo

• Faster production

• In-vivo production (eg viral injection)

• CRISPR (non viral) CART

• Lower cost – academic

Novel Targets

• Non-BCMA targets eg GPRC5d, CD70

• Dual targets eg. CD19 + BCMA, GPRC5d+ BCMA

Tumor Intrinsic

• Extramedullary Disease

• ISS3 –B2M

• High risk genomics

• High disease bulk/soluble blood Ag

• High soluble antigen in blood

• Functional ag loss

Conclusions

T cell Health

• better health in NDMM pts

• At apheresis - more naïve CD4+ T cells & less Tem

• higher CAR+ CD8+ stem cell like & lower CAR+ CD4r+ treg

• more T cell checkpoint expression after bsab

Microenvironment

• Higher inflammation/cytopenias correlate with worse CART outcomes

• Higher S100 (associated with MDSC) correlate with worse outcomes with CARTs and bispecifics

• S100 promotes T cell exhuaustion, T reg differentiation

Acknowledgements: Multiple Myeloma Team at UCSF

Patients and Caregivers

MDs

• Shagun Arora

• Ajai Chari

• Alfred Chung

• Anupama Kumar

• Tom Martin

• Nina Shah

• Peter Sayre

• Jeff Wolf

NPs/PAs

• Helen Diiulio

• Grace Sevilla

• Sam Shenoy (R)

• Nancy Wong

• Laura Zitella

Cellular Therapy Staff

• Jennifer Knoche

• Gabbi Perez

• Cheryl Slagle

RNs

• Jennifer Brustein

• Lisa Dunn

• Katharina Ganapathi

• Julie Mccluggage (R)

• Jenner Wells

• Jeani Wilmoth (R)

• Samanha Zylberman (R)

Social Workers

• Nina Balsamo

• Isabel Curtin

• Rachel Dornhelm

• Judy Smoker

• Ana Zermeño

Dieticians

Medical Assistants

• Nashia Raley

• Joan Viyela

Pharmacists

• Richard Fong

• Mimi Lo Administrative Staff

• Shelby Abuhatzira

• Deza Lynn Villanueva

Data Analyst

• Vishakha Malhotra

Clinical Research Staff (R)

• Jungeun Kim

• Alicia Aschauer

• Mrugakshi Dave

• Edlimar Delgado

• Liam Gima-Lange

• Kenya Gomez

• Jenny Nguyen

• Lauren Nguyen

• Ruixin Sun

• Lena Truong

Translational Research Team

Julia Carnevale

• Nupura Kale

• Stefanie Bachl

• Carter Ching

Justin Eyquem

 Chang Liu

 Joseph Muldoon

 Alexis Talbot

David Oh

• Kai Wu Arun Wiita

 Nikhil Chilakapati

 Szu-Ying Chen

 Bonell Patiño Escobar

 Haley Johnson

 Corynn Kasap,

 Adila Izgutdina

Thank you to

2025 IMWG Summit Sponsors

Working Group Meetings

Smoldering Multiple Myeloma – Dorado Room, Level -1 West Wing

Immune Therapy – Stay in this room

Quality of Life – Polar Room, -2 West Wing

Bone Disease – Alfa Room, - 2 North Wing

Combined Mass Spectrometry and MRD – Orione Room, - 2 West Wing

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.
2025 IMWG Summit Slides Tuesday by International Myeloma Foundation - Issuu