An IgG1-like bispecific antibody targeting CD52 and CD20 for the treatment of B-cell malignancies

Methods ◽  
2019 ◽  
Vol 154 ◽  
pp. 70-76 ◽  
Author(s):  
Junpeng Qi ◽  
Shih-Shih Chen ◽  
Nicholas Chiorazzi ◽  
Christoph Rader
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4170-4170
Author(s):  
Simone C. Oostindie ◽  
Hilma J. Van Der Horst ◽  
Marije B. Overdijk ◽  
Kristin Strumane ◽  
Sandra Verploegen ◽  
...  

Abstract CD37 is a tetraspanin plasma membrane protein abundantly expressed on B-cells and represents a promising therapeutic target for the treatment of B-cell malignancies. Although complement-dependent cytotoxicity (CDC) has proven to be a powerful Fc-mediated effector function for killing hematological cancer cells, CD37 antibody-based therapeutics currently in clinical development are poor inducers of CDC. Here we present DuoHexaBody-CD37, a novel humanized IgG1 bispecific antibody targeting two different CD37 epitopes, with an E430G hexamerization-enhancing mutation, for the potential treatment of B-cell malignancies. The natural process of antibody hexamer formation through intermolecular Fc-Fc interactions between IgG molecules after cell surface antigen binding can be improved by introducing a single point mutation such as E430G in the IgG Fc domain, thereby facilitating more efficient C1q binding and complement activation (Diebolder et al., Science 2014; de Jong et al., PLoS Biol 2016). The hexamerization-enhancing mutation E430G was introduced into two humanized CD37 monoclonal antibodies (mAbs) that bind non-overlapping CD37 epitopes. Different antibody formats and combinations, including the single antibodies, combinations of the mAbs and bispecific mAbs were tested for their capacity to induce CDC and antibody-dependent cellular cytotoxicity (ADCC). The bispecific hexamerization-enhanced antibody variant DuoHexaBody-CD37, showed superior CDC activity compared to the single hexamerization-enhanced mAbs and the combination thereof, both in vitro over a range of different B-cell lines, and ex vivo in tumor cell samples obtained from patients with chronic lymphocytic leukemia (CLL). In a CDC assay using tumor cells obtained from a relapsed/refractory CLL patient who received prior treatment with rituximab, ibrutinib and idelalisib, DuoHexaBody-CD37 induced almost complete lysis (84% lysis at a concentration 100 µg/mL), thereby outperforming the single HexaBody molecules (15% and 23% lysis) and the combination (57%) (Figure 1). In addition to its potent CDC activity, DuoHexaBody-CD37 was also capable of inducing potent ADCC of Daudi cells (EC50 = 12.3 ± 9.5 ng/mL), as assessed using peripheral blood mononuclear cells from 8 healthy human donors in a standard chromium release assay. In assays using whole blood from 6 healthy human donors, DuoHexaBody-CD37 showed efficient B-cell binding and potent and specific depletion of the B-cell population (98% ± 1.3% depletion at 10 µg/mL, EC50 = 0.85 ± 0.284 µg/mL). Furthermore, DuoHexaBody-CD37 induced significant inhibition of tumor growth in vivo in Daudi-luc Burkitt's lymphoma and JVM-3 CLL mouse xenograft models, at doses as low as 0.1 and 1 mg/kg (p<0.05), respectively. In summary, we present a novel therapeutic antibody that, for the first time, combines proprietary DuoBody® and HexaBody® platforms. DuoHexaBody-CD37 induced highly potent CDC and efficient ADCC in preclinical models, suggesting that DuoHexaBody-CD37 may serve as a potential therapeutic mAb for the treatment of human B-cell malignancies. Disclosures Oostindie: Genmab: Employment, Equity Ownership. Van Der Horst:Genmab: Research Funding. Overdijk:Genmab: Employment, Equity Ownership. Strumane:Genmab: Employment, Equity Ownership. Verploegen:Genmab: Employment, Equity Ownership. Lindorfer:Genmab: Research Funding. Cook:Genmab: Research Funding. Chamuleau:Gilead: Research Funding; BMS: Research Funding; celgene: Research Funding; Genmab: Research Funding. Mutis:Gilead: Research Funding; Celgene: Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Genmab: Research Funding; Novartis: Research Funding; OnkImmune: Research Funding. Schuurman:Genmab: Employment, Other: Warrants. Sasser:Genmab: Employment, Equity Ownership. Taylor:Genmab: Research Funding. Parren:Genmab: Equity Ownership; Lava Therapeutics: Employment. Beurskens:Genmab: Employment, Equity Ownership. Breij:Genmab: Employment, Equity Ownership.


Antibodies ◽  
2013 ◽  
Vol 2 (4) ◽  
pp. 338-352 ◽  
Author(s):  
Sandra Lüttgau ◽  
Dorothée Deppe ◽  
Saskia Meyer ◽  
Regina Fertig ◽  
Hossein Panjideh ◽  
...  

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 8065-8065 ◽  
Author(s):  
Trishna Goswami ◽  
Andres Forero ◽  
Mehdi Hamadani ◽  
Anne Sonet ◽  
Gregor Verhoef ◽  
...  

8065 Background: Novel B-cell targeting agents, including monoclonal antibodies such as rituximab, are among recent advances in treatment of B-cell malignancies. New approaches are needed for patients progressing after rituximab-based therapies. MEDI-551 is an afucosylated monoclonal antibody targeting CD-19, a B-cell restricted transmembrane protein with enhanced affinity and antibody-dependent cellular cytotoxicity. Methods: Pts with relapsed or refractory follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia, or multiple myeloma received single agent MEDI-551 at dosages ranging from 0.5 mg/kg to 12 mg/kg via intravenous infusion over 28-day cycles; cohorts 1-6 received 0.5, 1, 2, 4, 8, and 12 mg/kg, respectively. Results: 25 pts were enrolled in the phase I portion Jun 2010–Aug 2011. No maximum tolerated dose (MTD) was achieved. Most AEs were grade 1/2 with dose-independent frequency and severity (Table). Six pts had grade 3 toxicities including tumor lysis syndrome, infusion reaction, thrombocytopenia, and neutropenia, or grade 4 neutropenia. No grade 5 AEs were seen. All pts recovered. Three partial responses (PR) and 2 complete responses (CR) were seen in DLBCL and FL pts at 0.5, 4, and 8 mg/kg. Activity included a CR lasting 9 mo. in a FL pt in cohort 1, who is currently being retreated with MEDI-551 on relapse. Conclusions: MEDI-551 demonstrated a safety profile warranting further study and showed no MTD reached at the highest dose studied. Anti-tumor activity is suggested by the responses achieved across dose levels. Phase II is currently enrolling subjects. This study is funded by MedImmune, LLC. [Table: see text]


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1670-1670
Author(s):  
Takahiro Ohashi ◽  
Hitoshi Miyashita ◽  
Yuko Nagata ◽  
Satoko Tatebe ◽  
Hayuma Otsuka ◽  
...  

Abstract Introduction: IgM composes a part of the B cell receptor, and its expression is restricted to B cell lineage cells. There have been several reports that refractory B cell malignant cells frequently express IgM. For example, activated B cell-like (ABC) diffuse large cell lymphoma (DLBCL) is a subtype with a poor prognosis following R-CHOP treatment (Lenz et al., N Engl J Med, 2008), and the tumor cells generally express IgM-type immunoglobulin (Ruminy et al., Leukemia, 2011). In addition, IgM-expressing chronic lymphocytic leukemia (CLL) was also found to have a poor prognosis (D'Avola et al., Blood, 2016). Therefore, IgM is a promising therapeutic target for refractory B cell malignancies. However, anti-IgM antibodies may not function fully in the body due to the presence of soluble IgM. To improve the selectivity to the IgM expressing cells, we produced a bispecific antibody bound to IgM and HLA-DR, which are both expressed by B cells, and it was able to bind IgM+HLA-DR+ cells in the presence of soluble IgM. Thus, the aim of the present study was to examine the in vitro anti-tumor activity of the anti-IgM/HLA-DR bispecific antibody against B cell lymphoma, and to clarify its efficacy and tolerability in non-human primates (NHP). Methods: BTA106 is the bispecific antibody that can bind to IgM and HLA-DR, and BTA111 is glycoengineered BTA106. To investigate their direct inhibitory effects and Fc-mediated effects against B cell lymphoma cell lines, a growth inhibition assay, antibody-dependent cell cytotoxicity (ADCC) assay and complement dependent cytotoxicity (CDC) assay were performed. In the growth inhibition assay and CDC assay, cell proliferation was measured by WST-8. In the ADCC assay, human peripheral blood mononuclear cells were used as effector cells. Dead cells were stained with propidium iodide and measured by flow cytometry. JeKo-1 (Mantle cell lymphoma, MCL), Ramos (Burkitt's lymphoma, BL), RL (DLBCL), B104 (B cell lymphoma) and two rituximab-resistant cell lines, RL-4RH (Czuczman et al., Clin Cancer Res., 2008) and RRBL1 (generously provided by Dr. Tomita, Tomita et al., Int J Hematol., 2007), were used. To clarify the efficacy and tolerability of BTA106 in NHP, a dose escalation study and single-dose administration study were performed using cynomolgus monkeys. Results: BTA106 and BTA111 demonstrated direct growth inhibition activity, and CDC and ADCC activity, although their intensities differed among cell lines. In the case of rituximab-resistant cell lines, RRBL1 and RL-4RH, BTA106 and BTA111 exhibited weak or no growth inhibitory effects, but their CDC and ADCC activity was superior to that of rituximab. Development of resistance to rituximab is also a poor prognostic factor and remains problematic. Thus, BTA106 and BTA111 are expected as therapeutic antibodies to overcome rituximab resistance. A dose escalation study and single dose study were performed in cynomolgus monkeys. BTA106 greatly reduced peripheral B cells. Moreover, after the administration of BTA106, lymph follicles disappeared in axillary lymph nodes. In these studies, decreased locomotor activity and vomiting were observed; however, these symptoms resolved 2 hrs after injection. Other BTA106-related clinical toxicities or changes in body weight were not observed. Conclusions: In the in vitro studies, BTA106 and BTA111 demonstrated significant anti-tumor effects on rituximab-sensitive and -resistant B cell lymphoma cell lines. The efficacy and toxicity studies in cynomolgus monkeys revealed that BTA106 depletes peripheral and lymph node B cells, and it was well tolerated. BTA111 was considered to be significantly more effective than BTA106 due to glycoengineering, which induced high ADCC activity. These data suggest that BTA111 is an effective and potent therapeutic antibody for B cell malignancies, especially rituximab-resistant B cell lymphoma. Currently, a repeated administration study of BTA111 with cynomolgus monkeys to evaluate its efficacy and toxicity is ongoing. Disclosures Ohashi: Zenyaku Kogyo Co., Ltd.: Employment. Miyashita:Zenyaku Kogyo Co., Ltd.: Employment. Nagata:Zenyaku Kogyo Co., Ltd.: Employment. Tatebe:Zenyaku Kogyo Co., Ltd.: Employment. Otsuka:Zenyaku Kogyo Co., Ltd.: Employment. Suzuki:Zenyaku Kogyo Co., Ltd.: Employment. Sasaki:Zenyaku Kogyo Co., Ltd.: Employment. Enami:Zenyaku Kogyo Co., Ltd.: Employment. Tsukada:Zenyaku Kogyo Co., Ltd.: Employment. Fukushima:Zenyaku Kogyo Co., Ltd.: Employment.


2017 ◽  
Author(s):  
Krzysztof Masternak ◽  
Xavier Chauchet ◽  
Vanessa Buatois ◽  
Susana Salgado-Pires ◽  
Limin Shang ◽  
...  

Blood ◽  
2018 ◽  
Vol 131 (6) ◽  
pp. 611-620 ◽  
Author(s):  
Damian J. Green ◽  
Shyril O’Steen ◽  
Yukang Lin ◽  
Melissa L. Comstock ◽  
Aimee L. Kenoyer ◽  
...  

Key Points A novel bispecific antibody against CD38 eradicates MM and NHL tumors in murine models. CD38-bispecific antibody pretargeting addresses limitations associated with radioimmunotherapy.


2020 ◽  
Vol 9 (1) ◽  
pp. 50-61
Author(s):  
Iris de Weerdt ◽  
Roeland Lameris ◽  
George L. Scheffer ◽  
Jana Vree ◽  
Renate de Boer ◽  
...  

2015 ◽  
Vol 7 (287) ◽  
pp. 287ra70-287ra70 ◽  
Author(s):  
Liping L. Sun ◽  
Diego Ellerman ◽  
Mary Mathieu ◽  
Maria Hristopoulos ◽  
Xiaocheng Chen ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document