-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

4645 The Differential Effects of Bridging Therapy on Immune Microenvironment and Outcomes with Idecabtagene-Vicleucel in Multiple Myeloma

Program: Oral and Poster Abstracts
Session: 651. Multiple Myeloma and Plasma Cell Dyscrasias: Basic and Translational: Poster III
Hematology Disease Topics & Pathways:
Research, Adult, Translational Research, Chimeric Antigen Receptor (CAR)-T Cell Therapies, Plasma Cell Disorders, Diseases, Immune mechanism, Biological therapies, Treatment Considerations, Lymphoid Malignancies, Computational biology, Biological Processes, Study Population, Human
Monday, December 9, 2024, 6:00 PM-8:00 PM

Praneeth Reddy Sudalagunta, PhD1*, David R. Noyes, MS2*, Gabriel De Avila3*, Alexandra Achille2*, Rafael Renatino-Canevarolo, PhD1*, Xiaohong Zhao, MD, PhD2*, Maria Silva1*, Angel Perez1*, Mark B. Meads, PhD4*, Lauren C. Peres, PhD, MPH5*, Doris K. Hansen, MD6*, Kenneth H. Shain, MD, PhD4 and Ariosto Siqueira Silva, PhD7*

1Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
2Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
3H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
4H. Lee Moffitt Cancer Center, Tampa, FL
5Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL
6Moffitt Cancer Center, Tampa, FL
7Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL

Introduction: T cell engaging immunotherapies have shown great promise in treating patients with relapsed/refractory multiple myeloma (RRMM). In this study, we investigated the role of bridging therapy on immune composition and immune-tumor microenvironment (iTME), and its subsequent impact on outcomes of patients with RRMM treated with commercial idecabtagene-vicleucel (ide-cel).

Methods: This study included 171 patients with RRMM patients (baseline characteristics similar to Afrough et.al., Blood Cancer Journal, 2024) treated with ide-cel from May 25, 2021, to November 2, 2023, at Moffitt Cancer Center. Differences in progression-free survival (PFS) were compared either by choice of bridging therapy or immune markers using Kaplan-Meier survival curves, log-rank tests, and Cox proportional hazard regression models. Absolute lymphocyte counts (ALC) were obtained for all patients at apheresis and pre-LD (lymphodepletion) to determine the role of bridging on ALC and the impact of ALC on outcomes to ide-cel. A representative sub-sampling of 39 MM patients’ iTME (CD138-negative selection from bone marrow mononuclear cells) post-apheresis/bridging but pre-LD were characterized by multi-parameter flow cytometry (MPF) using a 36-parameter lymphoid panel on Cytek Aurora from viably frozen samples and a 21-parameter myeloid panel on BD Symphony A5 within 24 hours of sample collection and analyzed using FlowJo V10 software.

Results: Ide-cel treated patients with RRMM were categorized into seven mutually exclusive groups by choice of bridging therapy, where BTZ (bortezomib) bridging (n=14) had significantly shorter PFS compared to no bridging (n = 64) group by log-rank test p = 0.03 and HR = 4.69 [1.39 – 15.84] (consistent with Afrough et.al.), while CFZ (carfilzomib)/SELI (n = 2, p = 0.62, HR = 2.81 [0.38 – 20.71]), BTZ/SELI (n = 8, p = 0.39, HR = 2.07 [0.61 – 6.99]), CFZ (n = 26, p = 0.33, HR = 1.65 [0.72 – 3.77]), SELI other (n = 5, p = 0.89, HR = 1.23 [0.26 – 5.93]), and other bridging (n = 52, p = 0.89, HR = 1.00 [0.55 – 1.81]; includes IMIDs, monoclonal antibodies, DCEP, venetoclax, etc.) groups showed non-significant differences in PFS when compared to no bridging.

ALC measurements were compared between apheresis and pre-LD, where patients receiving a BTZ-based regimen showed a significant decrease in ALC (paired t-test, p = 0.03), while patients receiving any other bridging therapy (p = 0.94) or no bridging (p = 0.83) showed no significant differences. A comparison of PFS between ALC high (top 30 percentile) and ALC low (bottom 30 percentile) pre-LD showed that the ALC low group had significantly lower PFS (log-rank test p = 0.03 and HR = 2.06 [1.11 – 3.80]), suggesting that the decrease in ALC associated with BTZ bridging may lead to shorter PFS.

Flow characterization of iTME post-bridging/pre-LD of 39 patients revealed that a lower CD4:CD8 ratio (bottom 10th percentile vs remaining) was associated with shorter PFS (log-rank test p = 0.06 and HR = 16.67 [1.79 – 155.56]). An unpaired t-test comparing the CD4:CD8 ratio between no bridging (median = 1.00) and BTZ (median = 0.38) groups showed a significant decrease (p = 0.005) in CD4:CD8 ratio, while no significant differences were observed in other groups; although BTZ/SELI (median = 0.59) and SELI other (median = 1.07) groups had improved CD4:CD8 ratios compared to BTZ bridging group.

Lastly, expression of T cell inhibitory markers 2B4 on CD4+ and CD160 on CD8+ T cells were significantly higher in BTZ (p = 0.007 and 0.01, respectively) bridging group compared to no bridging, while comparisons with other groups showed no significant differences. A comparison of PFS between the top 10 percentile and rest of the cohort by expression of 2B4 on CD4+ and CD160 on CD8+ showed an association with shorter PFS (p = 0.095 and HR = 10.61 [1.3 – 86.7], p=0.003 and HR = 52.9 [5.8 – 480.9], respectively).

Conclusion: BTZ bridging-associated shorter PFS following ide-cel treatment was attributed to therapy-induced changes to immune function, while other bridging therapies (SELI, CFZ, immunologics, etc.) showed non-significant changes. Intriguingly, SELI associated modulation of the T cell repertoire was able to rescue the negative effects of BTZ on ide-cel outcomes in SELI/BTZ group. The observed BTZ (but not CFZ) dependent impacts on iTME likely stem from differences in mechanisms of action and drug pharmacokinetics and need further investigation in a larger patient cohort.

Disclosures: Sudalagunta: FORUS Therapeutics: Honoraria. Peres: Karyopharm Therapeutics: Research Funding; Bristol Myers Squibb: Research Funding. Shain: BMS: Consultancy, Research Funding; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Research Funding; Adaptive Biotech: Consultancy; Karyopharm, Janssen, Adaptive Biotechnologies, GlaxoSmithKline, BMS, Sanofi, and Regeneron: Honoraria; Sanofi: Consultancy; Takeda: Consultancy; Abbvie: Research Funding; Amgen: Research Funding; Glaxo Smith Kline: Consultancy, Membership on an entity's Board of Directors or advisory committees. Siqueira Silva: KARYOPHARM: Research Funding; ABBVIE: Research Funding.

*signifies non-member of ASH