Abstract
Introduction Immunotherapy has shown clinical promise in multiple myeloma (MM), but relapse remains common, largely due to the immunosuppressive bone marrow (BM) microenvironment, characterized by T cell exhaustion and the accumulation of myeloid-derived suppressor cells (MDSCs). Recent advances highlight the therapeutic promise of 4-1BB (CD137) agonist antibodies in enhancing T cell–mediated anti-tumor responses, though their efficacy in MM remains poorly defined. Investigating 4-1BB expression dynamics in a preclinical MM model informed our rationale for targeting this pathway. Tasquinimod (TasQ), a small-molecule immunomodulatory agent, currently evaluated in a phase Ib/IIa clinical trial in MM patients (NCT04405167), offers a complementary strategy. By inhibiting the S100A9 signaling pathway, TasQ interferes with the recruitment and function of MDSCs, resulting in a less suppressive tumor microenvironment (TME) which reestablishes the anti -tumor immunity. In this study, we assessed the therapeutic potential of two distinct 4-1BB agonist antibody clones in an immunocompetent preclinical MM model. Building on these findings, we further explored the combination of the most effective 4-1BB agonist with TasQ, aiming to overcome the immunosuppressive tumor microenvironment, and strengthen anti-myeloma immune activity.
Methods 4-1BB expression was analyzed during disease progression in 5T33MM mice using single-cell RNA sequencing of spleen and BM, 2 myeloma-infiltrating organs. These findings were validated by flow cytometry in both 5T33MM and 5TGM1 models. Therapeutic potential was assessed by treating 5TGM1 tumor-bearing mice with two 4-1BB agonist clones—LOB12.3 (IgG1κ; n=7/group) and 3H3 (IgG2a; n=5/group), using clone-specific isotype controls. Beginning on day 3 post-tumor inoculation, mice received 100µg of antibody intraperitoneally twice weekly until end stage. In a follow-up experiment, the lead 4-1BB agonist was combined with TasQ, administered at 30mg/kg in drinking water, to evaluate the impact of dual targeting the immunosuppressive TME (n=11/group). Tumor burden was assessed by determining the percentage of plasmacytosis in BM and spleen through cytospin stainings, together with M-protein measurement by serum electrophoresis. Immunomodulating effects were investigated using multi-parameter flow cytometry. Statistical significance was determined using the Mann–Whitney U test or one-way ANOVA, with p<0.05 considered significant.
Results 4-1BB was predominantly detected on T cells and natural killer (NK) cells, with its expression further increasing as the disease progressed. Treatment of 5TGM1 mice with 4-1BB agonists significantly increased the percentage of CD4+ and CD8+ T cells in the BM and spleen. Interestingly, treatment with clone LOB12.3 resulted in a significant reduction in NK cell percentages in both the BM and spleen, while clone 3H3 selectively reduced splenic NK cells. Therapeutically, clone 3H3 significantly decreased M-protein levels and BM plasmacytosis (p<0.01), while no significant effects were observed for clone LOB12.3.
Treatment with the IgG2a-formatted 4-1BB agonist combined with TasQ led to a significant reduction in M-protein levels and BM plasmacytosis (p<0001). Plasmacytosis was 62.50% in the isotype control group, decreased to 36.18% with 4-1BB agonist, 37.64% with TasQ, and further reduced to 14.09% in the combination therapy group, highlighting the enhanced efficacy of dual treatment. These effects were mediated by increased granzyme B–mediated T and NK-cell activation and enhanced differentiation of effector T cells (CD44⁺CD62L⁻). While this therapy did not alter the frequency of dendritic cells (DCs) in the BM, it enhanced their maturation, as evidenced by increased CD86 expression—particularly on type 1 and type 2 conventional DC subsets.
Conclusion In conclusion, our findings demonstrate that 4-1BB activation can enhance anti-tumor immunity in MM; however, the therapeutic efficacy is dependent on the specific agonist used. Notably, the IgG2a-formatted 4-1BB agonist showed superior activity, underscoring the importance of isotype selection in achieving optimal immunotherapeutic outcomes. Moreover, co-administration with TasQ enhanced therapeutic efficacy, supporting the potential benefit of a combinatorial approach. Further investigation is warranted to elucidate the mechanisms driving these responses and to refine 4-1BB-targeted strategies for effective clinical translation.
This feature is available to Subscribers Only
Sign In or Create an Account Close Modal