| Peer-Reviewed

Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma

Received: 28 April 2021    Accepted: 15 May 2021    Published: 26 May 2021
Views:       Downloads:
Abstract

Multiple myeloma (MM) is a malignancy characterized by abnormal proliferation of clonal plasma cells, and it is the second most common hematologic malignancy in the world after non-Hodgkin lymphoma. In recent years, significant progress has been made in the clinical treatment of MM. In particular, certain novel drugs, such as bortezomib, lenalidomide, and carfilzomib, have greatly improved the survival rate of patients with MM. However, because of drug resistance, most MM patients eventually suffer a relapse and die of the disease. In this study, the chimeric antigen receptor-modified T cell (CAR-T cell) technology, which has achieved success in recent clinical trials for B-cell acute lymphoblastic leukemia (B-ALL), was used. In view of the high CD138 expression in MM cells and the presence of the CD138–/CD19+ phenotype in a small subset of MM cells, and based on preliminary findings of effective killing of MM cells by CD19-CAR-T cells in clinical studies, CD138- and CD19-directed CAR-T cells were constructed. Through in vitro experiments and the use of a mouse model, we proved that these two types of CAR-T cells possess strong biological activity in the specific killing of target cells, and that the concomitant use of these cells significantly enhances the killing effect in an MM mouse model.

Published in Journal of Cancer Treatment and Research (Volume 9, Issue 1)
DOI 10.11648/j.jctr.20210901.12
Page(s) 10-21
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Multiple Myeloma, CAR-T, CD138 (syndecan-1), CD19

References
[1] Eshhar Z, Waks T, Bendavid A, et al. Functional expression of chimeric receptor genes in human T-cells. J Immunol Methods 2001; 248: 67–76.
[2] Hegde M, Corder A, Chow KK, et al. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T-cells in glioblastoma. Mol Ther 2013; 21: 2087–2101.
[3] Irving BA, Weiss A. The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways. Cell 1991; 64: 891–901.
[4] Sadelain M, Riviere I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer 2003; 3: 35–45.
[5] Brocker T, Karjalainen K. Signals through T cell receptor-zeta chain alone are in sufficient to prime resting T lymphocytes. J Exp Med 1995; 181: 1653–1659.
[6] Grada Z, Hegde M, Byrd T, et al. TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy. Mol Ther Nucleic Acids 2013; 2: e105.
[7] Tamada, K, Geng, D, Sakoda, Y, et al. Redirecting gene-modified T-cells toward various cancer types using tagged antibodies. Clin Cancer Res. 2013; 18: 6436–6445.
[8] Urbanska K, Lanitis E, Poussin M, et al. A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor. Cancer Res 2012; 72: 1844–1852.
[9] Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2016; 6: 224ra25.
[10] Lee DW, Barrett DM, Mackall C, Orentas R, Grupp SA. The future is now: chimeric antigen receptors as new targeted therapies for childhood cancer. Clin Cancer Res 2012; 18: 2780–2790.
[11] Han EQ, Li X, Wang C, Li T, Han S. Chimeric antigen receptor-engineered T cells for cancer immunotherapy: progress and challenges. J Hematol Oncol 2013; 6: 47.
[12] Chilosi M, Adami F, Lestani M, et al. CD138/syndecan-1: a useful immunohistochemical marker of normal and neoplastic plasma cells on routine trephine bone marrow biopsies. Mod Pathol 1999; 12: 1101–1106.
[13] Lin P, Owens R, Tricot G, Wilson CS. Flow Cytometric Immunophenotypic Analysis of 306 Cases of Multiple Myeloma. American Journal of Clinical Pathology 2004; 121: 482–488.
[14] Wei A, Juneja S. Bone marrow immunohistology of plasma cell neoplasms. J Clin Pathol 2003, 406–411.
[15] Wijdenes J, Vooijs WC, Clement C, et al. A plasmocyte selective monoclonal antibody (B-B4) recognizes syndecan-1. Br J Haematol 1996; 94: 318–323.
[16] Wijdenes J, Clement C, Klein B, et al. Leukocyte Typing VI: White Cell Differentiation Antigens. New York 1998.
[17] Lamorte S, Ferrero S, Aschero S, Monitillo L, Bussolati B, Omede P, et al. Syndecan-1 promotes the angiogenic phenotype of multiple myeloma endothelial cells. Leukemia 2012; 26: 1081–1090.
[18] Witzig TE, Kimlinger T, Stenson M, Therneau T. Syndecan-1 expression on malignant cells from the blood and marrow of patients with plasma cell proliferative disorders and B-cell chronic lymphocytic leukemia. Leukaemia & Lymphoma 1998; 31: 167–175.
[19] Yang Y, Yaccoby S, Liu W, Langford JK, Pumphrey CY, et al. Soluble syndecan-1 promotes growth of myeloma tumors in vivo. Blood 2002; 100: 610–617.
[20] Jiang H, Zhang WH, Shang P, Zhang H, et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Molecular oncology 2014; 8: 297-310.
[21] Guo B, Chen M, Han Q, Hui F, Dai H, Zhang W, et al. CD138-directed adoptive immunotherapy of chimeric antigen receptor (CAR)-modified T cells for multiple myeloma. J Cell Immunother 2016; 2: 28–35.
[22] Boucher K, Parquet N, Widen R, Shain K, Baz R, et al. Stemness of B cell progenitors in multiple myeloma bone marrow. Clin Cancer Res 2012; 18: 6155–6168.
[23] Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood 2004; 103: 2332-2336.
[24] Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res 2008; 68: 190–197.
[25] Chaidos A, Barnes CP, Cowan G, May PC, Melo V, Hatjiharissi E, et al. Clinical drug resistance linked to interconvertible phenotypic and functional states of tumor-propagating cells in multiple myeloma. Blood 2013; 121: 318–328.
[26] Yaccoby S. The phenotypic plasticity of myeloma plasma cells as expressed by dedifferentiation into an immature, resilient, and apoptosis- resistant phenotype. Clin Cancer Res 2005; 11: 7599-7606.
[27] Masahiro A, Takeshi H, Toshio M. Concise review: Defining and targeting myeloma stem cell-like cells. Stem cells 2014; 32: 1067-1073.
[28] Garfall AL, Maus MV, Hwang WT, et al. Chimeric Antigen Receptor T-cells against CD19 for Multiple Myeloma. N Engl J Med 2015; 373: 1040–1047.
[29] You FT, Jiang LC, et al. Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified Anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci 2016; 59: 386–397.
[30] Chen Y, You FT, et al. Gene-modified NK-92MI cells expressing a chimeric CD16-BB-ζ or CD64-BB-ζ receptor exhibit enhanced cancer-killing ability in combination with therapeutic antibody. Oncotarget 2017; 8: 37128-37139.
[31] Lecoeur H, Fevrier M, Garcia S, Riviere Y, Gougeon ML. A novel flow cytometric assay for quantitation and multiparametric characterization of cell-mediated cytotoxicity. J Immunol Methods 2001; 253: 177–187.
[32] Textor A, Listopad JJ, Wuhrman LL, Perez C, Kruschinski A, Chmielewski M, et al. Efficacy of CAR T-cell therapy in large tumors relies upon stromal targeting by IFN gamma. Cancer Res 2014; 74: 6796– 6805.
[33] Long AH, Haso WM, et al. 4-1BB Costimulation Ameliorates T Cell Exhaustion Induced by Tonic Signaling of Chimeric Antigen Receptors. Nat Med 2015; 21: 581-590.
[34] Chmielewski M, Hombach AA, Abken H. Of CARs and TRUCKs: chimeric antigen receptor (CAR) T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunol Rev 2014; 257: 83–90.
[35] Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 2016; 16: 566–581.
[36] Lima M, Teixeira MA, Fonseca S, et al. Immunophenotypic aberrations, DNA content, and cell cycle analysis of plasma cells in patients with myeloma and monoclonal gammopathies. Blood Cells Mol Dis 2000; 26: 634-645.
[37] Rousseau C, Ferrer L, Supiot S, Bardies M, Davodeau F, et al. Dosimetry results suggest feasibility of radioimmunotherapy using anti-CD138 (B-B4) antibody in multiple myeloma patients. Tumour Biol 2012; 33: 679-688.
[38] Jagannath S, Heffner T, Avigan D, Lutz R, Osterroth F, et al. BT062, An Antibody-Drug Conjugate Directed Against CD138, Shows Clinical Activity in Patients with Relapsed or Relapsed/Refractory Multiple Myeloma. Blood 2011; 118: Abstract 305.
[39] Robert O, Carpenter, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res 2013; 19: 2048-2060.
[40] Novak AJ, Darce JR, Arendt BK, et al. Expression of BCMA, TACI, and BAFF-R in multiple myeloma: a mechanism for growth and survival. Blood 2004; 103: 689-694.
[41] Ryan MC, Hering M, Peckham D, et al. Antibody targeting of B-cell maturation antigen on malignant plasma cells. Mol Cancer Ther 2007; 6: 3009-3018.
[42] Ali SA, Shi V, Maric I, et al. T cells expressing an anti-B-cell-maturation-antigen chimeric antigen re-ceptor cause remissions of multiple myeloma. Blood 2016; 128: 1688-1700.
[43] Berdeja JG, Lin Y, Raje N, Siegel D, et al. Clinical remissions and limited toxicity in a first-in-human multicenter study of bb2121, a novel anti-BCMA CAR T cell therapy for relapsed/refractory multiple myeloma. Annual Meeting of the EORTC/NCI/AACR. Munich, Germany 2016.
[44] Cohen AD, Garfall AL, Stadtmauer EA, Lacey SF, Lancaster E, et al. B-cell maturation antigen (BCMA)-specific chimeric antigen receptor T cells (CART-BCMA) for multiple myeloma (MM): initial safety and efficacy from a phase I study. Blood 2016; 128: 1147.
[45] Garfall AL, Lancaster E, Stadtmauer EA, Lacey SF, Dengel K, Ambrose DE, et al. Posterior reversible encephalopathy syndrome (PRES) after infusion of anti-BCMA CAR T cells (CART-BCMA) for multiple myeloma: successful treatment with cyclophospha- mide. Blood 2016; 128: 5702.
[46] Maria O, Felipe B, Matthew JF, Marcela VM. CARs in the lead against multiple myeloma. Curr Hematol Malig Rep 2017; 12: 119-125.
[47] Abate Daga D, Lagisetty K, Tran E, Zheng Z, Gattinoni L, Yu Z, et al. A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. Hum Gene Ther 2014; 25: 1003–1012.
[48] Hombach A, Rappl G, Abken H. Arming cytokine-induced killer cells with chimeric antigen receptors: CD28 outperforms combined CD28-OX40 "super-stimulation". Mol Ther 2013; 21: 2268–2277.
[49] Gupta R, Bhaskar A, Kumar L, Sharma A, Jain P. Flow cytometric immunophenotyping and minimal residual disease analysis in multiple myeloma. American Journal of Clinical Pathology. 2009; 132: 728–732.
Cite This Article
  • APA Style

    Songbo Zhao, Chao Wu, Jialu Li, Yafen Li, Gangli An, et al. (2021). Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma. Journal of Cancer Treatment and Research, 9(1), 10-21. https://doi.org/10.11648/j.jctr.20210901.12

    Copy | Download

    ACS Style

    Songbo Zhao; Chao Wu; Jialu Li; Yafen Li; Gangli An, et al. Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma. J. Cancer Treat. Res. 2021, 9(1), 10-21. doi: 10.11648/j.jctr.20210901.12

    Copy | Download

    AMA Style

    Songbo Zhao, Chao Wu, Jialu Li, Yafen Li, Gangli An, et al. Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma. J Cancer Treat Res. 2021;9(1):10-21. doi: 10.11648/j.jctr.20210901.12

    Copy | Download

  • @article{10.11648/j.jctr.20210901.12,
      author = {Songbo Zhao and Chao Wu and Jialu Li and Yafen Li and Gangli An and Huimin Meng and Zixuan Li and Lin Yang},
      title = {Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma},
      journal = {Journal of Cancer Treatment and Research},
      volume = {9},
      number = {1},
      pages = {10-21},
      doi = {10.11648/j.jctr.20210901.12},
      url = {https://doi.org/10.11648/j.jctr.20210901.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.jctr.20210901.12},
      abstract = {Multiple myeloma (MM) is a malignancy characterized by abnormal proliferation of clonal plasma cells, and it is the second most common hematologic malignancy in the world after non-Hodgkin lymphoma. In recent years, significant progress has been made in the clinical treatment of MM. In particular, certain novel drugs, such as bortezomib, lenalidomide, and carfilzomib, have greatly improved the survival rate of patients with MM. However, because of drug resistance, most MM patients eventually suffer a relapse and die of the disease. In this study, the chimeric antigen receptor-modified T cell (CAR-T cell) technology, which has achieved success in recent clinical trials for B-cell acute lymphoblastic leukemia (B-ALL), was used. In view of the high CD138 expression in MM cells and the presence of the CD138–/CD19+ phenotype in a small subset of MM cells, and based on preliminary findings of effective killing of MM cells by CD19-CAR-T cells in clinical studies, CD138- and CD19-directed CAR-T cells were constructed. Through in vitro experiments and the use of a mouse model, we proved that these two types of CAR-T cells possess strong biological activity in the specific killing of target cells, and that the concomitant use of these cells significantly enhances the killing effect in an MM mouse model.},
     year = {2021}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Concomitant Use of CD138- and CD19-directed Chimeric Antigen Receptor-modified T Cells Enhances Cytotoxicity Towards Multiple Myeloma
    AU  - Songbo Zhao
    AU  - Chao Wu
    AU  - Jialu Li
    AU  - Yafen Li
    AU  - Gangli An
    AU  - Huimin Meng
    AU  - Zixuan Li
    AU  - Lin Yang
    Y1  - 2021/05/26
    PY  - 2021
    N1  - https://doi.org/10.11648/j.jctr.20210901.12
    DO  - 10.11648/j.jctr.20210901.12
    T2  - Journal of Cancer Treatment and Research
    JF  - Journal of Cancer Treatment and Research
    JO  - Journal of Cancer Treatment and Research
    SP  - 10
    EP  - 21
    PB  - Science Publishing Group
    SN  - 2376-7790
    UR  - https://doi.org/10.11648/j.jctr.20210901.12
    AB  - Multiple myeloma (MM) is a malignancy characterized by abnormal proliferation of clonal plasma cells, and it is the second most common hematologic malignancy in the world after non-Hodgkin lymphoma. In recent years, significant progress has been made in the clinical treatment of MM. In particular, certain novel drugs, such as bortezomib, lenalidomide, and carfilzomib, have greatly improved the survival rate of patients with MM. However, because of drug resistance, most MM patients eventually suffer a relapse and die of the disease. In this study, the chimeric antigen receptor-modified T cell (CAR-T cell) technology, which has achieved success in recent clinical trials for B-cell acute lymphoblastic leukemia (B-ALL), was used. In view of the high CD138 expression in MM cells and the presence of the CD138–/CD19+ phenotype in a small subset of MM cells, and based on preliminary findings of effective killing of MM cells by CD19-CAR-T cells in clinical studies, CD138- and CD19-directed CAR-T cells were constructed. Through in vitro experiments and the use of a mouse model, we proved that these two types of CAR-T cells possess strong biological activity in the specific killing of target cells, and that the concomitant use of these cells significantly enhances the killing effect in an MM mouse model.
    VL  - 9
    IS  - 1
    ER  - 

    Copy | Download

Author Information
  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • Persongen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • The Cyrus Tang Hematology Center, Soochow University, Suzhou, PR China

  • Sections