scispace - formally typeset
Search or ask a question
Journal ArticleDOI

SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies.

TL;DR: These results validate CD38 as a therapeutic target and support the current evaluation of this unique CD38-targeting functional antibody in phase I clinical trials in patients with CD38+ B-cell malignancies.
Abstract: Purpose: The CD38 cell surface antigen is expressed in diverse hematologic malignancies including multiple myeloma, B-cell non-Hodgkin lymphoma (NHL), B-cell chronic lymphocytic leukemia, B-cell acute lymphoblastic leukemia (ALL), and T-cell ALL. Here, we assessed the antitumor activity of the anti-CD38 antibody SAR650984. Experimental Design: Activity of SAR650984 was examined on lymphoma, leukemia and multiple myeloma cell lines, primary multiple myeloma samples, and multiple myeloma xenograft models in immunodeficient mice. Results: We identified a humanized anti-CD38 antibody with strong proapoptotic activity independent of cross-linking agents, and potent effector functions including complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and antibody-dependent cellular phagocytosis (ADCP), equivalent in vitro to rituximab in CD20 + and CD38 + models. This unique antibody, termed SAR650984, inhibited the ADP-ribosyl cyclase activity of CD38, likely through an allosteric antagonism as suggested by 3D structure analysis of the complex. In vivo , SAR650984 was active in diverse NHL, ALL, and multiple myeloma CD38 + tumor xenograft models. SAR650984 demonstrated single-agent activity comparable with rituximab or cyclophosphamide in Daudi or SU-DHL-8 lymphoma xenograft models with induction of the proapoptotic marker cleaved capase-7. In addition, SAR650984 had more potent antitumor activity than bortezomib in NCI-H929 and Molp-8 multiple myeloma xenograft studies. Consistent with its mode of action, SAR650984 demonstrated potent proapoptotic activity against CD38 + human primary multiple myeloma cells. Conclusion: These results validate CD38 as a therapeutic target and support the current evaluation of this unique CD38-targeting functional antibody in phase I clinical trials in patients with CD38 + B-cell malignancies. Clin Cancer Res; 20(17); 4574–83. ©2014 AACR .
Citations
More filters
Journal ArticleDOI
TL;DR: Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma, and no maximum tolerated dose was identified in part 1.
Abstract: BACKGROUND Multiple myeloma cells uniformly overexpress CD38. We studied daratumumab, a CD38-targeting, human IgG1κ monoclonal antibody, in a phase 1–2 trial involving patients with relapsed myeloma or relapsed myeloma that was refractory to two or more prior lines of therapy. METHODS In part 1, the dose-escalation phase, we administered daratumumab at doses of 0.005 to 24 mg per kilogram of body weight. In part 2, the dose-expansion phase, 30 patients received 8 mg per kilogram of daratumumab and 42 received 16 mg per kilogram, administered once weekly (8 doses), twice monthly (8 doses), and monthly for up to 24 months. End points included safety, efficacy, and pharmacokinetics. RESULTS No maximum tolerated dose was identified in part 1. In part 2, the median time since diagnosis was 5.7 years. Patients had received a median of four prior treatments; 79% of the patients had disease that was refractory to the last therapy received (64% had disease refractory to proteasome inhibitors and immunomodulatory drugs and 64% had disease refractory to bortezomib and lenalidomide), and 76% had received autologous stem-cell transplants. Infusion-related reactions in part 2 were mild (71% of patients had an event of any grade, and 1% had an event of grade 3), with no dose-dependent adverse events. The most common adverse events of grade 3 or 4 (in ≥5% of patients) were pneumonia and thrombocytope nia. The overall response rate was 36% in the cohort that received 16 mg per kilogram (15 patients had a partial response or better, including 2 with a complete response and 2 with a very good partial response) and 10% in the cohort that received 8 mg per kilogram (3 had a partial response). In the cohort that received 16 mg per kilogram, the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 8.1), and 65% (95% CI, 28 to 86) of the patients who had a response did not have progression at 12 months. CONCLUSIONS Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma. (Funded by Janssen Research and Development and Genmab; ClinicalTrials.gov number, NCT00574288.)

882 citations

Journal ArticleDOI
11 Mar 2015-mAbs
TL;DR: It is demonstrated that phagocytosis is a fast, potent and clinically relevant mechanism of action that may contribute to the therapeutic activity of DARA in multiple myeloma and potentially other hematological tumors.
Abstract: Daratumumab (DARA) is a human CD38-specific IgG1 antibody that is in clinical development for the treatment of multiple myeloma (MM). The potential for IgG1 antibodies to induce macrophage-mediated phagocytosis, in combination with the known presence of macrophages in the tumor microenvironment in MM and other hematological tumors, led us to investigate the contribution of antibody-dependent, macrophage-mediated phagocytosis to DARA's mechanism of action. Live cell imaging revealed that DARA efficiently induced macrophage-mediated phagocytosis, in which individual macrophages rapidly and sequentially engulfed multiple tumor cells. DARA-dependent phagocytosis by mouse and human macrophages was also observed in an in vitro flow cytometry assay, using a range of MM and Burkitt's lymphoma cell lines. Phagocytosis contributed to DARA's anti-tumor activity in vivo, in both a subcutaneous and an intravenous leukemic xenograft mouse model. Finally, DARA was shown to induce macrophage-mediated phagocytosis of MM c...

387 citations

Journal ArticleDOI
Michel Attal, Paul G. Richardson1, S. Vincent Rajkumar2, Jesús F. San-Miguel3, Meral Beksac4, Ivan Spicka5, Xavier Leleu, Fredrik Schjesvold6, Fredrik Schjesvold7, Philippe Moreau, Meletios A. Dimopoulos8, Jeffrey Shang Yi Huang9, Jiri Minarik10, Michele Cavo11, H. Miles Prince12, Sandrine Macé, Kathryn P. Corzo13, Frank Campana13, Solenn Le-Guennec, Franck Dubin, Kenneth C. Anderson1, Vincent Rajkumar2, Simon J. Harrison, Wojt Janowski, Ian Kerridge, Andrew Spencer, Michel Delforge, Karel Fostier, Philip Vlummens, Ka Lung Wu, Richard Leblanc, Michel Pavic, Michael Sebag, Roman Hájek, Vladimir Maisnar, Ludek Pour, Henrik Gregersen, Lotfi Benbouker, Denis Caillot, Martine Escoffre-Barbe, Thierry Facon, Laurent Frenzel, Cyrille Hulin, Lionel Karlin, Brigitte Kolb, Brigitte Pegourie, Aurore Perrot, Mourad Tiab, Laure Vincent, Dietger Niederwieser, Achilles Anagnostopoulos, Sosana Delimpasi, Marie Christine Kyrtsonis, Anargyros Symeonidis, Árpád Illés, Gabor Mikala, Zsolt Nagy, Sara Bringen, Paolo Corradini, Ciceri Fabio, Roberto M. Lemoli, Anna Marina Liberati, Chiara Nozzoli, Renato Zambello, Shinsuke Iida, Takashi Ikeda, Satoshi Iyama, Morio Matsumoto, Chihiro Shimazaki, Kazutaka Sunami, Kenshi Suzuki, Michihiro Uchiyama, Youngil Koh, Ki-Hyun Kim, Jae Hoon Lee, Chang-Ki Min, Hillary Blacklock, Hugh Goodman, AJ Neylon, David Simpson, Sebastian Grosicki, Artur Jurczyszyn, Adam Walter-Croneck, Krzysztof Warzocha, Luis Araujo, Claudia Moreira, Vadim A Doronin, Larisa P. Mendeleeva, Vladimir I. Vorobyev, Andrej Vranovsky, Adrian Alegre, Mercedes Gironella, Marta Sonia Gonzalez Perez, Carmen Montes, Enrique M. Ocio, Paula Rodriguez, Mats Hardling, Birgitta Lauri, Ming Chung Wang, Su Peng Yeh, Mutlu Arat, Fatih Demirkan, Zafer Gulbas, Sevgi Kalayoglu Besisik, Ihsan Karadogan, Tulin Tuglular, Ali Ünal, Filiz Vural, Jonathan Sive, Matthew Streetly, Kwee Yong, Jason Tache 
TL;DR: The aim of this study was to determine the progression-free survival benefit of isatuximab plus pomalidomide and dexamethasone compared with pomidine and dexAMethas one in patients with relapsed and refractory multiple myeloma.

328 citations

Journal ArticleDOI
04 Jan 2018-Blood
TL;DR: Deep responses and prolonged progression-free survival can be achieved in relapsed/refractory MM patients when CD38 antibodies are combined with immunomodulatory agents or proteasome inhibitors, and CD38-targeting antibodies are generally well tolerated and induce partial response or better in heavily pretreated MM patients as monotherapy.

307 citations

Journal ArticleDOI
TL;DR: CD38‐targeting antibodies have a favorable toxicity profile in patients, and early clinical data show a marked activity in MM, while studies in other hematological malignancies are ongoing.
Abstract: CD38 is a multifunctional cell surface protein that has receptor as well as enzyme functions. The protein is generally expressed at low levels on various hematological and solid tissues, while plasma cells express particularly high levels of CD38. The protein is also expressed in a subset of hematological tumors, and shows especially broad and high expression levels in plasma cell tumors such as multiple myeloma (MM). Together, this triggered the development of various therapeutic CD38 antibodies, including daratumumab, isatuximab, and MOR202. Daratumumab binds a unique CD38 epitope and showed strong anti-tumor activity in preclinical models. The antibody engages diverse mechanisms of action, including complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, programmed cell death, modulation of enzymatic activity, and immunomodulatory activity. CD38-targeting antibodies have a favorable toxicity profile in patients, and early clinical data show a marked activity in MM, while studies in other hematological malignancies are ongoing. Daratumumab has single agent activity and a limited toxicity profile, allowing favorable combination therapies with existing as well as emerging therapies, which are currently evaluated in the clinic. Finally, CD38 antibodies may have a role in the treatment of diseases beyond hematological malignancies, including solid tumors and antibody-mediated autoimmune diseases.

271 citations


Cites background from "SAR650984, a novel humanized CD38-t..."

  • ...Lammerts van Bueren J, et al. Direct in vitro comparison of daratumumab with surrogate analogs of CD38 antibodies MOR03087, SAR650984 and Ab79....

    [...]

  • ...Martin TG, Strickland SA, Glenn M, Zheng W, Daskalakis N, Mikhael JR. SAR650984, a CD38 monoclonal antibody in patients with selected CD38+ hematological malignancies- data from a dose-escalation phase I study....

    [...]

  • ...Isatuximab is currently being tested in early clinical trials in CD38-positive hematological malignancies, including MM (64, 65)....

    [...]

  • ...Similarly, isatuximab kills lymphoma cells via ADCC and ADCP, and also has strong proapoptotic activity in lymphoma in the absence of crosslinking agents (64)....

    [...]

  • ...Nevertheless, daratumumab and isatuximab showed a marked anti-tumor activity in mouse xenograft models of NHL in vivo (63, 64, 134)....

    [...]

References
More filters
Journal ArticleDOI
TL;DR: Select IgG1 variants with improved binding to FcγRIIIA exhibited up to 100% enhancement in antibody-dependent cell cytotoxicity using human effector cells; these variants included changes at residues not found at the binding interface in the IgG/Fcγ RIIIA co-crystal structure.

1,788 citations

Journal ArticleDOI
TL;DR: It is suggested that is it now possible to define stages of T-cell differentiation that can be related to T- cell malignancies in humans.
Abstract: A series of monoclonal antibodies was used to define three discrete stages of human intrathymic T-cell differentiation The earliest stage was confined to <10% of thymocytes, which were·reactive with both OKT9 and OKT10 Subsequently, approximately 70% of human thymocytes acquired a thymocyte-restricted antigen, OKT6, lost OKT9 antigen, and expressed reactivity with OKT4 and OKT5 These last two monoclonal antibodies were previously shown to define inducer (helper) and cytotoxic/suppressor populations, respectively, in peripheral blood The OKT4+, OKT5+, OKT6+ “common” thymocyte population represents the majority of thymocytes and accounts for more than 70% of thymocytes With further maturation, thymocytes lose OKT6 reactivity, segregate into OKT4+ and OKT5+ subsets, and acquire reactivity with OKT3 (and OKT1) This latter stage corresponds to the more functionally mature subset The possible relationship of acute lymphoblastic leukemia of T-cell lineage to these proposed stages of intrathymic differentiation was determined Analysis of 25 tumor populations showed that 21 could be related to one or another differentiative stage The majority (15/21) were derived from an early thymocyte or prothymocyte subpopulation, 5/25 were derived from a common thymocyte subpopulation, and 1/25 was derived from a mature (OKT3+) subpopulation These data suggest that is it now possible to define stages of T-cell differentiation that can be related to T-cell malignancies in humans

1,578 citations

Journal ArticleDOI
15 Mar 1991-Blood
TL;DR: Direct evidence is provided for the existence of a single class of progenitors with extensive proliferative capacity in human BM and an experimental approach for their purification, manipulation, and further characterization is provided.

924 citations

Journal ArticleDOI
03 Jun 2010-Blood
TL;DR: In human lymphoma xenograft models, GA101 exhibits superior antitumor activity, resulting in the induction of complete tumor remission and increased overall survival and in nonhuman primates, GA 101 demonstrates superior B cell-depleting activity in lymphoid tissue, including in lymph nodes and spleen.

841 citations


"SAR650984, a novel humanized CD38-t..." refers background in this paper

  • ...This type of enhanced proapoptotic activity has been described for type II anti-CD20 antibodies such as tositumumab or obinutuzumab (37)....

    [...]

Related Papers (5)