Elsevier

Cellular Immunology

Volume 345, November 2019, 103964
Cellular Immunology

The promise of chimeric antigen receptor (CAR) T cell therapy in multiple myeloma

https://doi.org/10.1016/j.cellimm.2019.103964Get rights and content

Highlights

  • Multiple myeloma remains an incurable disease.

  • BCMA CAR T therapy shows promise in trials of relapsed and/or refractory myeloma.

  • Timing, targets, toxicities, and resistance mechanisms of CAR T need further studies.

Abstract

A cure for multiple myeloma (MM), a malignancy of plasma cells, remains elusive. Nearly all myeloma patients will eventually relapse and develop resistance to currently available treatments. There is an unmet medical need to develop novel and effective therapies that can induce sustained responses. Early phase clinical trials using chimeric antigen receptor (CAR) T cell therapy have shown great promise in the treatment of relapsed and/or refractory MM. In this review article, we provide an overview of the CAR constructs, the gene transfer vector systems, and approaches for T cell activation and expansion. We then summarize the outcomes of several early phase clinical trials of CAR T cell therapy in MM and the novel CAR T targets that are under development. Finally, we explore the potential mechanisms that result in disease relapse after CAR T therapy and propose future directions in CAR T therapy in MM.

Section snippets

Multiple myeloma

Multiple Myeloma (MM) is a malignancy of terminally differentiated plasma cells typically characterized by clonal proliferation of these plasma cells in the bone marrow. MM represents 1% of all malignancies and 18% of hematologic malignancies in the United States; accounting for an estimated 32,110 new diagnoses and 12,960 deaths in 2019 alone [1]. The diagnosis of MM is made based on the presence of ≥10% clonal plasma cells in the bone marrow, and evidence of end organ damage (typically

Chimeric antigen receptor constructs

Cancer patients have profound defects in anti-tumor immunity, resulting in the failure to suppress tumor growth [22], [23]. The objective of CAR T therapy is to activate the anti-tumor immunity by adoptive infusion of a sufficient number of immune effector cells. Most of the immune effector cells used for the adoptive transfer are T cells, although NK cells have also been used. Autologous immune effector cells are typically used but allogeneic immune effector cells have been reported [24], [25]

Early phase multiple myeloma CAR T clinical trials targeting BCMA

B-cell maturation antigen (BCMA), also referred to as tumor necrosis factor receptor superfamily member 17 (TNFRSF17) or CD269, is the receptor for BAFF and APRIL and is expressed consistently on myeloma cells and normal plasma cells at varying intensities [54], [55], [56]. BCMA has been shown to promote multiple myeloma pathogenesis, and targeting BCMA has been shown to have potent anti-myeloma activity [56], [57], [58], [59]. BCMA antigen can be cleaved by gamma-secretase and released into

Phase I clinical trial with CAR T therapy targeting the κ light chain

Ramos et al. at the Baylor College of Medicine and Houston Methodist Hospital conducted a phase 1 clinical trial in patients with κ+ non-Hodgkin lymphoma/chronic lymphocytic leukemia or MM (ClinicalTrials.gov: NCT00881920) (Table 3) [74]. Autologous T cells were genetically engineered by retroviral vector to express a murine scFv fragment targeting the κ light chain of human immunoglobulin, a CD28 costimulatory domain and a CD3ζ domain. A spacer region derived from the human IgG1-CH2CH3 domains

CAR T therapy targeting SLAMF7

SLAMF7 (CD319, CS-1) is a member of the signaling lymphocytic activation molecule family of trans-membrane receptors. SLAMF7 is expressed on NK cells and a proportion of CD8+ T cells. SLAMF7 is uniformly expressed on malignant plasma cells in newly diagnosed multiple myeloma and in relapsed myeloma after intensive chemotherapy [86], [87], [88]. Furthermore, SLAMF7-CAR T cells were effective against myeloma cells in vitro and in vivo in a murine xenograft model [89]. Importantly, SLAMF7-CAR T

Mechanisms of resistance/relapse after CAR T therapy

Despite the promise of CAR T therapy in the treatment of MM, patients still relapse with CAR T therapy even after achieving MRD negativity. The progression-free survival in bb2121 and LCAR-B38M trial was 17.7 and 15 months, respectively. The mechanisms for the relapse remain to be characterized but may include the following.

Second CAR T therapy

At the 2019 ASCO annual meeting, Li et al. presented 4 cases who received human BCMA-targeting CAR T therapy (CT103A) after the patients’ myeloma relapsed from a murine BCMA-targeting CAR T therapy. The CT103A CAR construct is a lentiviral vector encoding a fully human scFV, CD8a hinger and transmembrane, 4-1BB costimulatory and CD3ζ activation domain. Patients received 1–6 × 106 CAR T cells/kg. All 4 patients had objective responses: 3 patients achieved a CR and 1 patient had a VGPR. More

Summary and future direction

CAR T therapy has shown promising results and safety profiles. It is highly likely that in the near future CAR T therapy will become an important modality of treatment especially for relapsed/refractory MM. It remains to be determined when will be the best time to incorporate CAR T therapy in MM: as part of induction therapy, in the relapsed setting, as an alternative to ASCT, or as an adjunctive to ASCT? Finally, the mechanisms of resistance/relapse with CAR T therapy remain to be

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgement

Funding: This work is supported by Duke Cancer Institute Fund, NIH R44CA199767, NIH 5T32 HL007057-42, NCI R21CA234701, and NIH R01CA197792.

Authors’ contributions

All authors wrote, read, and approved the final manuscript.

References (119)

  • J. Sheu et al.

    Large-scale production of lentiviral vector in a closed system hollow fiber bioreactor

    Mol. Ther. Meth. Clin. Dev.

    (2015)
  • A.-M. Rasmussen et al.

    Ex vivo expansion protocol for human tumor specific T cells for adoptive T cell therapy

    J. Immunol. Meth.

    (2010)
  • A.R. Medvec et al.

    Improved expansion and in vivo function of patient T cells by a serum-free medium

    Mol. Ther. Meth. Clin. Dev.

    (2018)
  • A.J. Novak et al.

    Expression of BCMA, TACI, and BAFF-R in multiple myeloma: a mechanism for growth and survival

    Blood

    (2004)
  • Y.-T. Tai et al.

    APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment

    Blood

    (2016)
  • Jennifer Brudno et al.

    T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor with a CD28 costimulatory moiety cause remissions of poor-prognosis relapsed multiple myeloma

    Blood

    (2017)
  • S.A. Ali et al.

    T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma

    Blood

    (2016)
  • E.L. Smith et al.

    Development and evaluation of an optimal human single-chain variable fragment-derived BCMA-targeted CAR T cell vector

    Mol. Ther.

    (2018)
  • Jesus G. Berdeja et al.

    Durable clinical responses in heavily pretreated patients with relapsed/refractory multiple myeloma: updated results from a multicenter study of bb2121 Anti-Bcma CAR T cell therapy

    Blood

    (2017)
  • B. Guo et al.

    CD138-directed adoptive immunotherapy of chimeric antigen receptor (CAR)-modified T cells for multiple myeloma

    J. Cell. Immunother.

    (2016)
  • Y.-T. Tai et al.

    Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu

    Blood

    (2008)
  • T. Gogishvili et al.

    SLAMF7-CAR T cells eliminate myeloma and confer selective fratricide of SLAMF7(+) normal lymphocytes

    Blood

    (2017)
  • L. Lee et al.

    An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma

    Blood

    (2018)
  • F. van Rhee et al.

    NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses

    Blood

    (2005)
  • P.G. Richardson et al.

    Management of treatment-emergent peripheral neuropathy in multiple myeloma

    Leukemia

    (2012)
  • N. Latov

    Pathogenesis and therapy of neuropathies associated with monoclonal gammopathies

    Ann. Neurol.

    (1995)
  • S.V. Rajkumar

    Multiple myeloma update on diagnosis, risk-stratification, and management

    Am. J. Hematol.

    (2016)
  • J. Mikhael et al.

    Treatment of multiple myeloma: ASCO and CCO joint clinical practice guideline

    J. Clin. Oncol.

    (2019)
  • P.G. Richardson et al.

    A phase 2 study of bortezomib in relapsed, refractory myeloma

    N. Engl. J. Med.

    (2003)
  • P.G. Richardson et al.

    Bortezomib or high-dose dexamethasone for relapsed multiple myeloma

    N. Engl. J. Med.

    (2005)
  • H. Ludwig et al.

    Survival and years of life lost in different age cohorts of patients with multiple myeloma

    J. Clin. Oncol.

    (2010)
  • A. Palumbo et al.

    Daratumumab, bortezomib, and dexamethasone for multiple myeloma

    N. Engl. J. Med.

    (2016)
  • M.A. Dimopoulos et al.

    Lenalidomidedaratumumab and dexamethasone for multiple myeloma

    N. Engl. J. Med.

    (2016)
  • H.M. Lokhorst et al.

    Targeting CD38 with daratumumab monotherapy in multiple myeloma

    N. Engl. J. Med.

    (2015)
  • S. Lonial et al.

    Elotuzumab therapy for relapsed or refractory multiple myeloma

    N. Engl. J. Med.

    (2015)
  • B. Barlogie et al.

    Incorporating bortezomib into upfront treatment for multiple myeloma: early results of total therapy 3

    Br. J. Haematol.

    (2007)
  • C.B. Reeder et al.

    Cyclophosphamide, bortezomib and dexamethasone induction for newly diagnosed multiple myeloma: high response rates in a phase II clinical trial

    Leukemia

    (2009)
  • S. Knop et al.

    Bortezomib, IV cyclophosphamide, and dexamethasone (VelCD) as induction therapy in newly diagnosed multiple myeloma: results of an interim analysis of the German DSMM Xia trial

    J. Clin. Oncol.

    (2009)
  • A. Palumbo et al.

    Bortezomib as induction before autologous transplantation, followed by lenalidomide as consolidation-maintenance in untreated multiple myeloma patients

    J. Clin. Oncol.

    (2010)
  • S.K. Kumar, I. Flinn, S.J. Noga, P. Hari, R. Rifkin, N. Callander, M. Bhandari, J.L. Wolf, C. Gasparetto, A. Krishnan,...
  • J. Carbone et al.

    Immune dysfunction in cancer patients

    Oncology (Huntingt)

    (2002)
  • G.P. Dunn et al.

    Cancer immunoediting: from immunosurveillance to tumor escape

    Nat. Immunol.

    (2002)
  • Y. Yang et al.

    Challenges and opportunities of allogeneic donor-derived CAR T cells

    Curr. Opin. Hematol.

    (2015)
  • A.T. Gacerez et al.

    How chimeric antigen receptor design affects adoptive T cell therapy

    J. Cell. Physiol.

    (2016)
  • X. Liu et al.

    Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice

    Cancer Res.

    (2015)
  • C. Wong et al.

    Human scFv antibody fragments specific for the epithelial tumour marker MUC-1, selected by phage display on living cells

    Cancer Immunol. Immunother.

    (2001)
  • C.M. Hammers et al.

    Antibody phage display: technique and applications

    J. Invest. Dermatol.

    (2014)
  • K.S. Kahlon et al.

    Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells

    Cancer Res.

    (2004)
  • D.M. Davies et al.

    Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells

    Mol. Med.

    (2012)
  • S. Guedan et al.

    Engineering and design of chimeric antigen receptors

    Mol. Ther. Meth. Clin. Dev.

    (2018)
  • Cited by (17)

    • PPAR agonists attenuate lenalidomide's anti-myeloma activity in vitro and in vivo

      2022, Cancer Letters
      Citation Excerpt :

      Lenalidomide is the most commonly used IMiD, and nearly all patients with MM receive it during the course of their treatment, especially during post-autologous stem cell transplant maintenance therapy. IMiDs bind to a shallow hydrophobic pocket of cereblon (CRBN), leading to the recruitment and targeted ubiquitination and degradation of IKAROS family zinc finger (IKZF)-1 and IKZF3 [7,8]. CRBN is critical for the anti-myeloma activity of IMiDs, and the expression level of CRBN is one of the major determining factors of drug sensitivity or resistance to IMiDs [9,10].

    • Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      The objective of this article is to provide a summary of the current status of MM CAR-T and offer potential future directions to optimize the utility of CAR T cell therapy in the treatment of MM. Although there are some differences in the details of each CAR construct, the most commonly used CARs are composed of five components, including 1) antigen-binding domain which is oftentimes single-chain variable fragment (scFv); 2) hinge (or spacer) which is connecting scFv and transmembrane domain; 3) transmembrane domain; 4) intracellular costimulatory domain; and 5) activation domain from CD3ζ (Feinberg et al., 2019; Gacerez et al., 2016). The scFv renders transduced T-cells to target the surface antigen of interest on cancer cells.

    • Current Status and Prospects of Immunotherapy for Osteosarcoma

      2022, Cancer Research on Prevention and Treatment
    View all citing articles on Scopus
    View full text