Abstract 4556: Preclinical pharmacology and safety of RO7297089, a novel anti-BCMA/CD16a bispecific antibody for the treatment of multiple myeloma

Author(s):  
Satoko Kakiuchi-Kiyota ◽  
Melissa M. Schutten ◽  
Adeyemi O. Adedeji ◽  
Hao Cai ◽  
Robert Hendricks ◽  
...  
Author(s):  
Sameer Quazi

Multiple Myeloma (MM) is one of the incurable types of cancer in plasma cells. While immense progress has been made in the treatment of this malignancy, a large percentage of patients were unable to adapt to such therapy. Additionally, these therapies might be associated with significant diseases and are not always tolerated well in all patients. Since cancer in plasma cells has no cure, patients develop resistance to treatments, resulting in R/R MM. BCMA is primarily produced on mature B cells. Its up-regulation and activation are associated with multiple myeloma in both murine and human models, indicating that this might be an effective therapeutic target for this type of malignancy. Additionally, BCMA's predictive value, association with effective clinical trials, and capacity to be utilized in previously difficult to observe patient populations, imply that it might be used as a biomarker for multiple myeloma. Numerous kinds of BCMA-targeting medicines have demonstrated antimyeloma efficacy in individuals with refractory/relapsed MM, including CAR T-cell treatments, ADCs, bispecific antibody constructs. Among these medications, CART cell-mediated BCMA therapy has shown significant outcomes in multiple myeloma clinical trials. This review article outlines CAR T cell mediated BCMA medicines have the efficiency to change the therapeutic pattern for multiple myeloma significantly.


eJHaem ◽  
2020 ◽  
Vol 1 (1) ◽  
pp. 113-121
Author(s):  
David M. Foureau ◽  
Manisha Bhutani ◽  
Myra Robinson ◽  
Fei Guo ◽  
Duy Pham ◽  
...  

2020 ◽  
Vol 4 (18) ◽  
pp. 4538-4549 ◽  
Author(s):  
Kodandaram Pillarisetti ◽  
Gordon Powers ◽  
Leopoldo Luistro ◽  
Alexander Babich ◽  
Eric Baldwin ◽  
...  

Abstract B-cell maturation antigen (BCMA), a member of the tumor necrosis factor family of receptors, is predominantly expressed on the surface of terminally differentiated B cells. BCMA is highly expressed on plasmablasts and plasma cells from multiple myeloma (MM) patient samples. We developed a BCMAxCD3 bispecific antibody (teclistamab [JNJ-64007957]) to recruit and activate T cells to kill BCMA-expressing MM cells. Teclistamab induced cytotoxicity of BCMA+ MM cell lines in vitro (H929 cells, 50% effective concentration [EC50] = 0.15 nM; MM.1R cells, EC50 = 0.06 nM; RPMI 8226 cells, EC50 = 0.45 nM) with concomitant T-cell activation (H929 cells, EC50 = 0.21 nM; MM.1R cells, EC50 = 0.1 nM; RPMI 8226 cells, EC50 = 0.28 nM) and cytokine release. This activity was further increased in the presence of a γ-secretase inhibitor (LY-411575). Teclistamab also depleted BCMA+ cells in bone marrow samples from MM patients in an ex vivo assay with an average EC50 value of 1.7 nM. Under more physiological conditions using healthy human whole blood, teclistamab mediated dose-dependent lysis of H929 cells and activation of T cells. Antitumor activity of teclistamab was also observed in 2 BCMA+ MM murine xenograft models inoculated with human T cells (tumor inhibition with H929 model and tumor regression with the RPMI 8226 model) compared with vehicle and antibody controls. The specific and potent activity of teclistamab against BCMA-expressing cells from MM cell lines, patient samples, and MM xenograft models warrant further evaluation of this bispecific antibody for the treatment of MM. Phase 1 clinical trials (monotherapy, #NCT03145181; combination therapy, #NCT04108195) are ongoing for patients with relapsed/refractory MM.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2752-2752
Author(s):  
Matthias Peipp ◽  
Claudia Ehlert ◽  
Matthias Staudinger ◽  
Joerg Bruenke ◽  
Georg Fey ◽  
...  

Abstract Different therapeutic options are available for the treatment of multiple myeloma patients, but conventional chemotherapy often is not able to completely eradicate the tumor. After high dose chemotherapy, complete remission with only minimal residual disease could be achieved in many patients, but additional targeted strategies may help to eradicate residual cells and improve prognosis. Therapy with monoclonal antibodies is well established in CD20 positive B-cell lymphomas, but in contrast not many suitable target antigens are defined / expressed by multiple myeloma cells. HM1.24 a surface molecule expressed on terminally differentiated B-lineage cells represents a promising candidate antigen that is overexpressed on multiple myeloma cells. Here the development of a recombinant bispecific single chain Fv HM1.24 × CD16 antibody (tandem format) with novel features is presented. The HM1.24×CD16 bispecific antibody was expressed in 293T cells and purified to homogeneity by two-step affinity chromatography. Binding to HM1.24 and CD16 was demonstrated by immunofluorescence staining and flow cytometry with antigen positive and negative cells. The lytic activity of the bispecific HM1.24×CD16 scFv was evaluated in an antibody-dependent cellular cytotoxicity (ADCC) assay with different myeloma cell lines (RPMI 8226, INA-6, U266, JK6L) and primary patient derived cells as targets. Mononuclear cells (MNC), isolated from healthy donors served as effector cells. The bispecific HM1.24×CD16 scFv mediated efficient lysis of all tested cell lines at concentrations as low as 1 nM. In direct comparison to an HM1.24-IgG1 control molecule, the recombinant bispecific antibody demonstrated superior lytic activity at saturating concentrations and showed significant enhanced killing capacity. In conclusion, the recombinant bispecific HM1.24×CD16 retained its antigen specificity and demonstrated efficient lytic activity against patient-derived tumor cell lines and primary material. These results indicate that the bispecific antibody may be promising as a new therapeutic strategy in multiple myeloma.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3176-3176 ◽  
Author(s):  
Dennis Cooper ◽  
Deepu Madduri ◽  
Suzanne Lentzsch ◽  
Sundar Jagannath ◽  
Jingjin Li ◽  
...  

Background Multiple myeloma (MM), the second most common hematologic malignancy, is characterized by the expansion of malignant plasma cells which express, the cell surface protein, B-cell maturation antigen (BCMA). Patients (pts) with advanced MM who are refractory to an immunomodulatory agent (IMiD), a proteasome inhibitor (PI), and an anti-CD38 monoclonal antibody (mAb) have an expected overall survival of <1 year (Gandhi et al. 2019). Given the therapeutic potential of utilizing BCMA to redirect T-cell effector function on multiple myeloma cells, we generated REGN5458, an anti-BCMA x anti-CD3 bispecific antibody that binds to both BCMA on plasma cells and to CD3 on T-cells. Here we describe the safety and clinical activity in relapsed/refractory MM patients treated on the initial dose level of REGN5458 in trial (NCT03761108). Methods The primary objectives of the Phase 1 portion of the study are to determine the safety, tolerability and occurrence of dose limiting toxicities (DLTs) of REGN5458. The primary objective of the Phase 2 portion is to assess the preliminary anti-tumor activity of REGN5458. Key secondary objectives include assessment of pharmacokinetics (PK) and pharmacodynamics. Eligible pts with MM must have >3 prior lines of therapy including a PI, IMiD and anti-CD38 antibody or progression on or after an anti-CD38 antibody and refractory to a PI and IMiD. Treatment consists of 16 weekly doses of REGN5458, followed by a maintenance phase of 12 doses administered every 2 weeks. Pts with progressive disease after initial response are eligible for retreatment. Response was assessed per the International Myeloma Working Group (IMWG) criteria. Results As of July 12, 2019, three pts have been treated at the initial dose level of 3 mg REGN5458. All pts had an ECOG score of 1. Pt 1, an 81-year-old male who had medullary plasmacytomas and cutaneous extramedullary plasmacytomas (EMPs), had received four prior lines of therapy. He experienced Grade (Gr) 1 cytokine release syndrome (CRS) that was treated with tocilizumab and corticosteroids because of persistent debilitating fever. This patient also experienced Gr 3 TEAEs including anemia, pain in both extremities (location of multiple sites of disease), and worsening hypertension within the DLT evaluation period. Subsequent to the DLT evaluation period, he had Gr 3 fatigue, Gr 3 febrile neutropenia, Gr 3 lung infection, Gr 3 atrial fibrillation, and Gr 4 septic shock. Pt 1 reached a partial response at Week 8 and a very good partial response (VGPR) as of Week 16 despite interruption of study drug at Week 14 due to TEAEs. This pt has IgG lambda myeloma and showed rapid decreases in both lambda free light chain and M-protein (Figure 1A) and resolution of medullary and cutaneous plasmacytomas following the Week 12 dose (Figure 1B and 1C). Pt 1 had transient cytokine elevations of interferon gamma, interleukin (IL)-6, and IL-10 following dosing through Week 5, consistent with mild CRS. Peripheral blood immune monitoring revealed increases in CD8 effector memory T-cells through Week 11, relative to other subsets which remained unchanged during the treatment period. Pt 1 remains in the treatment phase of the study. Pt 2 is a 76-year-old female who had received four prior lines of therapy and had extensive intra-abdominal EMPs. She had no ≥ Gr 2 TEAEs. Pt 2 had disease progression at first assessment and is in the follow-up phase of the study. Pt 3 is a 78-year-old female with seven prior lines of therapy. She experienced Gr 2 decreases in both platelets and neutrophils within the DLT evaluation period. Pt 3 had stable disease (SD) at first assessment and remains in the treatment phase of the study. No DLTs were reported. No pt experienced infusion-related reactions. No pt had Gr 5 TEAEs or discontinued treatment due to AEs. Additional PK and biomarker data will be presented. Conclusions/Summary In this FIH study of REGN5458, no DLTs were recorded in the first three pts treated with the initial dose. One pt responded with a VGPR and another had SD. The study is ongoing and recruiting pts at higher doses. Gandhi UH et al. Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia; 2019; DOI:10.1038/s41375-019-0435-7 Disclosures Madduri: Celgene: Consultancy; AbbVie: Consultancy; Foundation Medicine: Consultancy; Takeda: Consultancy. Lentzsch:Bayer: Consultancy; Columbia University: Patents & Royalties: 11-1F4mAb as Anti-Amyloid Strategy; Janssen: Consultancy; Caelum Biosciences: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Research Funding; Crossfires in hematologic Malignancies: Honoraria; International Myeloma Foundation: Honoraria; Multiple Myelopma Research Foundation: Honoraria; Abbvie: Consultancy; BMS: Consultancy; Proclara: Consultancy; Clinical Care Options: Speakers Bureau; Sanofi: Consultancy, Research Funding; Takeda: Consultancy. Jagannath:Celgene Corporation: Consultancy; Bristol-Myers Squibb: Consultancy; Merck & Co.: Consultancy; Janssen Pharmaceuticals: Consultancy; AbbVie: Consultancy; Karyopharm Therapeutics: Consultancy. Li:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Boyapati:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Adriaens:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Chokshi:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Zhu:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Lowy:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Weinreich:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Yancopoulos:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Sharma:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Karasarides:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Sternberg:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. OffLabel Disclosure: The data described in the abstract will report on use of REGN5458 in a first-in-human trial in patients with multiple myeloma.


2021 ◽  
pp. 1-4
Author(s):  
Madeeha Subhan Waleed ◽  

Multiple myeloma (MM), as defined by a clonal plasma cell proliferation, manifests as end organ damage caused by the abnormally high monoclonal paraprotein.In this article, we have reviewed the potential benefits of Bispecific antibodies (BsAbs) in MM patients. In addition, new BsAbs developments and clinical trials for various MM targets are discussed in detail. Bispecific antibodies are the types of antibodies that have two different antigen binding sites in one molecule. There are 100 different classes of BsAb and all these can be divided into 2 main categories based on their fragments and both categories are under trials for MM.Despite some studies showing adverse effects development of these new treatments is going to greatly contribute to improve outcomes for a wide group of patients which also requires further clinical studies to be conducted with focus on demonstration of efficacy and safety profile.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 8045-8045 ◽  
Author(s):  
Thorsten Gantke ◽  
Uwe Reusch ◽  
Christian Kellner ◽  
Kristina Ellwanger ◽  
Ivica Fucek ◽  
...  

8045 Background: Despite recent advances in the treatment of multiple myeloma (MM), novel therapies are needed to achieve long-lasting remissions in a greater number of patients. Natural killer (NK) cells play a key role in the immune response to MM and have been implicated in the clinical efficacy of current standard of care interventions, including IMiDs, proteasome inhibitors, recently approved immunotherapies and autologous stem cell transplantation (ASCT). Numerous strategies are being developed to enhance the natural NK-cell cytotoxicity against myeloma cells, which is frequently dysregulated in MM. Approaches include modulation of activity, through cytokine stimulation or immune checkpoint targeting, and adoptive transfer of culture expanded NK-cells in ASCT-eligible MM. While highly attractive, these approaches are non-targeted, as they rely on the natural cytotoxicity of NK-cells, and may benefit from antigen-specific retargeting and effector activation. AFM26 is a novel tetravalent, bispecific antibody designed to specifically enhance NK-cell anti-MM activity by redirecting NK-cell lysis to BCMA, an antigen expressed on MM cells. Methods: NK-cell engagement and cytotoxicity of AFM26 towards MM cell lines and freshly isolated tumor cells from MM patients was characterized in vitro and compared with classical antibody formats. Results: AFM26 engages NK-cells with superior avidity ( KD: 1-2nM) through bivalent interaction with CD16A (FcγRIIIa) and demonstrates extended cell surface retention that is not affected by high level IgG, as is particularly relevant in MM. Importantly, AFM26 does not induce NK-cell depletion but selectively induces potent and efficacious lysis of MM cells in vitro. Conclusions: In summary, AFM26 is a promising candidate to enhance NK-cell activity and confer tumor-specificity to NK-cells in MM. Differentiation of AFM26 from classical antibody formats and its potential for combination with cellular NK-cell therapies is highlighted.


2018 ◽  
Vol 36 (15_suppl) ◽  
pp. 8034-8034 ◽  
Author(s):  
Ben Buelow ◽  
Priya Choudry ◽  
Starlynn Clarke ◽  
Kevin Dang ◽  
Laura Davison ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document