Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2022

Open Access 01-12-2022 | Multiple Myeloma | Correspondence

FTL004, an anti-CD38 mAb with negligible RBC binding and enhanced pro-apoptotic activity, is a novel candidate for treatments of multiple myeloma and non-Hodgkin lymphoma

Authors: Guangbing Zhang, Cuiyu Guo, Yan Wang, Xianda Zhang, Shuang Liu, Wen Qu, Chunxia Chen, Lingli Yan, Zhouning Yang, Zhixiong Zhang, Xiaohua Jiang, Xiaofeng Chen, Hong Liu, Qinhuai Lai, Xian Wei, Ying Lu, Shengyan Zhao, Han Deng, Yuxi Wang, Lin Yu, Hongbin Yu, Yu Wu, Zhaoming Su, Pengyu Chen, Ziqing Ren, Meng Yu, Feng Qu, Yong Luo, Lantu Gou, Qing Li, Ying Huang, Fanxin Ma, Jinliang Yang

Published in: Journal of Hematology & Oncology | Issue 1/2022

Login to get access

Abstract

Anti-CD38 monoclonal antibodies (mAbs), daratumumab, and isatuximab have represented a breakthrough in the treatment of multiple myeloma (MM). Recently, CD38-based mAbs were expected to achieve increasing potential beyond MM, which encouraged us to develop new anti-CD38 mAbs to meet clinical needs. In this study, we developed a novel humanized anti-CD38 antibody, FTL004, which exhibited enhanced pro-apoptotic ability and negligible binding to red blood cells (RBCs). FTL004 presented a better ability to induce direct apoptosis independent of Fc-mediated cross-linking against lymphoma and MM cell lines as well as primary myeloma cells derived from MM patients. For instance, FTL004 induced RPMI 8226 cells with 55% early apoptosis cells compared with 20% in the isatuximab-treated group. Of interest, FTL004 showed ignorable binding to CD38 on human RBCs in contrast to tumor cells, even at concentrations up to 30 μg/mL. Furthermore, with an engineered Fc domain, FTL004 displayed stronger antibody-dependent cellular cytotoxicity (ADCC) against CD38+ malignant cells. In vivo MM and non-Hodgkin lymphoma tumor xenograft models showed that FTL004 possessed an effective anti-tumor effect. Cryo-electron microscopy structure resolved two epitope centers of FTL004 on CD38: one of which was unique while the other partly overlapped with that of isatuximab. Taken together, FTL004 distinguishes it from other CD38 targeting mAbs and represents a potential candidate for the treatment of MM and non-Hodgkin lymphoma.
Literature
1.
go back to reference Chini CCS, Peclat TR, Warner GM, Kashyap S, Espindola-Netto JM, de Oliveira GC, et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD(+) and NMN levels. Nat Metab. 2020;2(11):1284–304.CrossRef Chini CCS, Peclat TR, Warner GM, Kashyap S, Espindola-Netto JM, de Oliveira GC, et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD(+) and NMN levels. Nat Metab. 2020;2(11):1284–304.CrossRef
2.
go back to reference Mehta K, Shahid U, Malavasi F. Human CD38, a cell-surface protein with multiple functions. FASEB J Off Publ Fed Am Soc Exp Biol. 1996;10(12):1408–17. Mehta K, Shahid U, Malavasi F. Human CD38, a cell-surface protein with multiple functions. FASEB J Off Publ Fed Am Soc Exp Biol. 1996;10(12):1408–17.
3.
go back to reference van de Donk NW, Janmaat ML, Mutis T, van Lammerts Bueren JJ, Ahmadi T, Sasser AK, et al. Monoclonal antibodies targeting CD38 in hematological malignancies and beyond. Immunol Rev. 2016;270(1):95–112.CrossRef van de Donk NW, Janmaat ML, Mutis T, van Lammerts Bueren JJ, Ahmadi T, Sasser AK, et al. Monoclonal antibodies targeting CD38 in hematological malignancies and beyond. Immunol Rev. 2016;270(1):95–112.CrossRef
5.
go back to reference Ofran Y. Daratumumab: new indications revolving around “off-targets.” Haematologica. 2021;106(12):3032–3.CrossRef Ofran Y. Daratumumab: new indications revolving around “off-targets.” Haematologica. 2021;106(12):3032–3.CrossRef
6.
go back to reference Fedyk ER, Idamakanti N, Chen J, Estevam J, Palumbo A. The binding of CD38 therapeutics to red blood cells and platelets subverts depletion of target cells. Blood. 2019;134(Supplement_1):3136.CrossRef Fedyk ER, Idamakanti N, Chen J, Estevam J, Palumbo A. The binding of CD38 therapeutics to red blood cells and platelets subverts depletion of target cells. Blood. 2019;134(Supplement_1):3136.CrossRef
8.
go back to reference Lancman G, Arinsburg S, Jhang J, Cho HJ, Jagannath S, Madduri D, et al. Blood transfusion management for patients treated with anti-CD38 monoclonal antibodies. Front Immunol. 2018;9:2616.CrossRef Lancman G, Arinsburg S, Jhang J, Cho HJ, Jagannath S, Madduri D, et al. Blood transfusion management for patients treated with anti-CD38 monoclonal antibodies. Front Immunol. 2018;9:2616.CrossRef
9.
go back to reference Naeimi Kararoudi M, Nagai Y, Elmas E, de Souza Fernandes Pereira M, Ali SA, Imus PH, et al. CD38 deletion of human primary NK cells eliminates daratumumab-induced fratricide and boosts their effector activity. Blood. 2020;136(21):2416–27.CrossRef Naeimi Kararoudi M, Nagai Y, Elmas E, de Souza Fernandes Pereira M, Ali SA, Imus PH, et al. CD38 deletion of human primary NK cells eliminates daratumumab-induced fratricide and boosts their effector activity. Blood. 2020;136(21):2416–27.CrossRef
10.
go back to reference Jebamani P, Sriramulu DK, Jung ST, Lee S-G. Structural study on the impact of S239D/I332E mutations in the binding of Fc and FcγRIIIa. Biotechnol Bioprocess Eng. 2021;26(6):985–92.CrossRef Jebamani P, Sriramulu DK, Jung ST, Lee S-G. Structural study on the impact of S239D/I332E mutations in the binding of Fc and FcγRIIIa. Biotechnol Bioprocess Eng. 2021;26(6):985–92.CrossRef
11.
go back to reference Hong Y, Guo H, Wei M, Zhang Y, Fang M, Cheng T, et al. Cell-based reporter assays for measurements of antibody-mediated cellular cytotoxicity and phagocytosis against SARS-CoV-2 spike protein. J Virol Methods. 2022;307: 114564.CrossRef Hong Y, Guo H, Wei M, Zhang Y, Fang M, Cheng T, et al. Cell-based reporter assays for measurements of antibody-mediated cellular cytotoxicity and phagocytosis against SARS-CoV-2 spike protein. J Virol Methods. 2022;307: 114564.CrossRef
Metadata
Title
FTL004, an anti-CD38 mAb with negligible RBC binding and enhanced pro-apoptotic activity, is a novel candidate for treatments of multiple myeloma and non-Hodgkin lymphoma
Authors
Guangbing Zhang
Cuiyu Guo
Yan Wang
Xianda Zhang
Shuang Liu
Wen Qu
Chunxia Chen
Lingli Yan
Zhouning Yang
Zhixiong Zhang
Xiaohua Jiang
Xiaofeng Chen
Hong Liu
Qinhuai Lai
Xian Wei
Ying Lu
Shengyan Zhao
Han Deng
Yuxi Wang
Lin Yu
Hongbin Yu
Yu Wu
Zhaoming Su
Pengyu Chen
Ziqing Ren
Meng Yu
Feng Qu
Yong Luo
Lantu Gou
Qing Li
Ying Huang
Fanxin Ma
Jinliang Yang
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2022
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-022-01395-0

Other articles of this Issue 1/2022

Journal of Hematology & Oncology 1/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine