Elsevier

Blood Reviews

Volume 45, January 2021, 100695
Blood Reviews

Review
Revving the CAR – Combination strategies to enhance CAR T cell effectiveness

https://doi.org/10.1016/j.blre.2020.100695Get rights and content

Abstract

Chimeric antigen receptor (CAR) T cell therapy is currently approved for treatment of refractory B-cell malignancies. Response rates in these diseases are impressive by historical standards, but most patients do not have a durable response and there remains room for improvement. To date, CAR T cell activity has been even more limited in solid malignancies. These limitations are thought to be due to several pathways of resistance to CAR T cells, including cell-intrinsic mechanisms and the immunosuppressive tumor microenvironment. In this review, we discuss current experimental strategies that combine small molecules and monoclonal antibodies with CAR T cells to overcome these resistance mechanisms. We describe the biological rationale, pre-clinical data and clinical trials in progress that test the efficacy and safety of these combinations.

Introduction

Chimeric Antigen Receptor (CAR) T cell therapy has revolutionized the treatment of refractory B cell malignancies. It represents a new treatment paradigm, that leverages the ability of genetically modified T cells to seek and destroy tumor cells by identifying specific surface markers. The two products currently on the market are axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel), for the approved indications of relapsed or refractory pediatric B cell acute lymphoblastic leukemia (B-ALL) and aggressive B cell non-Hodgkin lymphoma (NHL). Despite the excitement surrounding CAR T cells and their promise, their full potential has yet to be realized. In pivotal trials of CAR T cell therapy for aggressive B cell lymphoma, the long term disease-free survival was between 30 and 40% [[1], [2], [3]]. In B-ALL 81% of patients had an initial response, with 59% of responders remaining relapse free beyond 1 year, bringing the rate of durable clinical benefit down to approximately half of the patients [4]. Though these results compare favorably to historical cohorts of heavily pretreated patients, there is significant room for improvement. In solid malignancies progress has been slower, owing both to the scarcity of tumor specific antigens, and a highly immunosuppressive tumor microenvironment (TME) [5]. There is intensive pre-clinical research and clinical trials underway to improve the efficacy of CAR T cells, focusing on recently described resistance mechanisms (summarized in Fig. 1). One approach is to use various small molecules and monoclonal antibodies in combination with CAR T cells to overcome tumor escape mechanisms and improve anti-tumor activity. In this review, we will focus on this strategy. We will detail the pre-clinical evidence to support combinations of certain drugs with CAR T cells, data from early phase trials in humans and ongoing trials.

Section snippets

Immunomodulatory drugs (IMiDs)

There is strong pre-clinical rationale to support combination of immunomodulatory drugs (IMiDs) with CAR T cells. IMiDs enhance T cell proliferation and cytokine production via an interleukin (IL)-2 dependent mechanism [6]. Thalidomide, pomalidomide and lenalidomide all directly bind to cereblon, the substrate recognition component of cullin-dependent ubiquitin ligase (CRL4CRBN) [7,8]. Upon binding, IMiDs enhance cereblon’s affinity towards certain substrates. IMiD treatment leads to enhanced

Conclusions and future directions

The early development efforts in CAR T cell therapy have focused on maximizing its single agent activity. Major advances have been made in optimizing various aspects CAR T cell therapy including LD chemotherapy, selection of target antigen, and the design of the CAR construct itself. However, despite significant successes, the true potential of CAR T cell therapy is yet to be realized. In particular, attempts to apply this strategy to solid tumors have encountered significant challenges. The

Practice points

  • New strategies are needed to improve the efficacy of CAR T cell therapy in hematological malignancies and extend their application to solid tumors.

  • Multiple mechanisms contribute to loss of CAR T cell activity. These mechanisms include, but are not limited to, exhaustion of infused T cells, loss of persistence, differentiation to effector phenotypes, defective immune synapses, downregulation of target antigens, and antigen heterogeneity.

  • Small molecules and antibodies currently in development may

Research Agenda

  • Designing early phase trials that combine CAR T cells with promising drugs currently in pre-clinical development including:

    • o

      Novel IMiDs

    • o

      SMAC mimetics

    • o

      Fas blockade

    • o

      IL-15 receptor agonists

    • o

      MDSC depleting agents

    • o

      AXL inhibitors

    • o

      Accelerators of reactive oxygen species.

  • Further optimizing dosing schedule of small molecules and antibodies to maximize efficacy

  • Developing strategies to mitigate additive and synergistic toxicities of combinatorial treatment.

Funding

We gratefully acknowledge funding from the Conquer Cancer Foundation (Young Investigator Award to RB) and the Margie and Andy Rooke Fund for Leukemia Research (to RR).

Declaration of competing interest

Ran Reshef receives research funding from Gilead, Incyte, Atara, Pharmacyclics, Shire, BMS, Takeda, and consulting fees from Gilead, Atara, Novartis, Magenta and BMS.

References (193)

  • J.A. Fraietta et al.

    Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia

    Blood

    (2016)
  • E. Ten Hacken et al.

    Microenvironment interactions and B-cell receptor signaling in chronic lymphocytic leukemia: implications for disease pathogenesis and treatment

    Biochim Biophys Acta

    (2016)
  • Q. Qiu et al.

    Targeted delivery of ibrutinib to tumor-associated macrophages by sialic acid-stearic acid conjugate modified nanocomplexes for cancer immunotherapy

    Acta Biomater

    (2019)
  • W.J. Norde et al.

    Coinhibitory molecules in hematologic malignancies: targets for therapeutic intervention

    Blood

    (2012)
  • E.S. Rijkers et al.

    The inhibitory CD200R is differentially expressed on human and mouse T and B lymphocytes

    Mol Immunol

    (2008)
  • J. Mock et al.

    Risk of major bleeding with ibrutinib

    Clin Lymphoma Myeloma Leuk

    (2018)
  • J.J. Shatzel et al.

    Ibrutinib-associated bleeding: pathogenesis, management and risk reduction strategies

    J Thromb Haemost

    (2017)
  • T. Gargett et al.

    GD2-specific CAR T cells undergo potent activation and deletion following antigen encounter but can be protected from activation-induced cell death by PD-1 blockade

    Mol Ther

    (2016)
  • S. Kleffel et al.

    Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth

    Cell.

    (2015)
  • S.N. Zolov et al.

    Programmed cell death protein 1 activation preferentially inhibits CD28.CAR-T cells

    Cytotherapy

    (2018)
  • E.A. Chong et al.

    PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: refueling the CAR

    Blood

    (2017)
  • A. Heczey et al.

    CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma

    Mol Ther

    (2017)
  • P. Kumar et al.

    A comprehensive review on the role of co-signaling receptors and Treg homeostasis in autoimmunity and tumor immunity

    J Autoimmun

    (2018)
  • M.C. Milone et al.

    Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo

    Mol Ther

    (2009)
  • S.S. Neelapu et al.

    Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma

    N Engl J Med

    (2017)
  • S.J. Schuster et al.

    Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma

    N Engl J Med

    (2019)
  • S.L. Maude et al.

    Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia

    N Engl J Med

    (2018)
  • P. Kosti et al.

    Perspectives on chimeric antigen receptor T-cell immunotherapy for solid tumors

    Front Immunol

    (2018)
  • P.H. Schafer et al.

    Enhancement of cytokine production and AP-1 transcriptional activity in T cells by thalidomide-related immunomodulatory drugs

    J Pharmacol Exp Ther

    (2003)
  • T. Ito et al.

    Identification of a primary target of thalidomide teratogenicity

    Science

    (2010)
  • A. Lopez-Girona et al.

    Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide

    Leukemia

    (2012)
  • A.K. Gandhi et al.

    Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.)

    Br J Haematol

    (2014)
  • G. Lu et al.

    The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins

    Science

    (2014)
  • S. O’Brien et al.

    Ikaros imposes a barrier to CD8+ T cell differentiation by restricting autocrine IL-2 production

    J Immunol

    (2014)
  • E.J. Kim et al.

    Enhanced immune-modulatory effects of thalidomide and dexamethasone co-treatment on T cell subsets

    Immunology

    (2017)
  • A.G. Ramsay et al.

    Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug

    J Clin Invest

    (2008)
  • A.J. Davenport et al.

    Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity

    Proc Natl Acad Sci U S A

    (2018)
  • K. Watanabe et al.

    Expanding the therapeutic window for CAR T cell therapy in solid tumors: the knowns and unknowns of CAR T cell biology

    Front Immunol

    (2018)
  • S. Kuramitsu et al.

    Lenalidomide enhances the function of chimeric antigen receptor T cells against the epidermal growth factor receptor variant III by enhancing immune synapses

    Cancer Gene Ther

    (2015)
  • R.L. Browning et al.

    Lenalidomide induces interleukin-21 production by T cells and enhances IL21-mediated cytotoxicity in chronic lymphocytic leukemia B cells

    Cancer Immunol Res

    (2016)
  • R. Loschinski et al.

    IL-21 modulates memory and exhaustion phenotype of T-cells in a fatty acid oxidation-dependent manner

    Oncotarget

    (2018)
  • H. Singh et al.

    Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies

    Cancer Res

    (2011)
  • P. Otahal et al.

    Lenalidomide enhances antitumor functions of chimeric antigen receptor modified T cells

    Oncoimmunology

    (2016)
  • G. Fouquet et al.

    Efficacy and safety profile of long-term exposure to lenalidomide in patients with recurrent multiple myeloma

    Cancer

    (2013)
  • S. Fried et al.

    Early and late hematologic toxicity following CD19 CAR-T cells

    Bone Marrow Transplant

    (2019)
  • X. Leleu et al.

    An international, multicenter, prospective, observational study of neutropenia in patients being treated with lenalidomide + dexamethasone for relapsed or relapsed/refractory multiple myeloma (RR-MM)

    Am J Hematol

    (2016)
  • S.S. Neelapu et al.

    A phase 2, open-label, multicenter study evaluating the safety and efficacy of axicabtagene ciloleucel in combination with either rituximab or lenalidomide in patients with refractory large B-cell lymphoma (ZUMA-14)

    Blood

    (2019)
  • S. Lonial et al.

    Translational and clinical evidence of a differentiated profile for the novel CELMoD, Iberdomide (CC-220)

    Blood

    (2019)
  • D.W. Rasco et al.

    A first-in-human study of novel cereblon modulator avadomide (CC-122) in advanced malignancies

    Clin Cancer Res

    (2019)
  • V. Werth et al.

    CT-07 A phase 2, randomised, placebo-controlled, double-blind, ascending dose study to evaluate efficacy, safety, and tolerability, pharmacokinetics, pharmacodynamics and pharmacogenetics of CC-220 in subjects with systemic lupus erythematosus

    Lupus Sci Med

    (2016)
  • Cited by (22)

    • Blasts in context: the impact of the immune environment on acute myeloid leukemia prognosis and treatment

      2023, Blood Reviews
      Citation Excerpt :

      These observations suggest a modulatory effect of the combination AZA/CAR on the immune environment of AML [119]. Other ways to overcome immune dysfunction and enhance adoptive T cell therapy were recently reviewed by our group and others [47,113,120,121]. Clinical translation of this approach is eagerly awaited and should include investigations on immune environment markers.

    • Metabolic and epigenetic orchestration of (CAR) T cell fate and function

      2022, Cancer Letters
      Citation Excerpt :

      The effectiveness of CAR T cells in solid tumors is also limited by several obstacles including i) impaired trafficking to, and penetration of, tumor sites; ii) antigen heterogeneity/loss; iii) immunosuppressive cells and inhibitory factors in the tumor milieu; iv) poor persistence, engraftment, and proliferation of adoptively transferred CAR T cells [7]. Combinatorial approaches including the addition of immune checkpoint blockers, oncolytic viruses, bispecific T cell engagers as well as cytokines that neutralize the suppressive environment have been tested pre-clinically, but there is currently no approved CAR T cell regimen for solid tumors [8–10]. As T cells lack endogenous fuel reserves, it is necessary to understand how local metabolism in the TME impedes CAR T cell function, and develop innovative solutions that reinforce metabolic and differentiation programs for optimal effectiveness.

    • Epigenetic strategies to boost CAR T cell therapy

      2021, Molecular Therapy
      Citation Excerpt :

      Combining CAR T cells with immune checkpoint blockades, oncolytic viruses, bispecific T cell engagers, and cytokines has increased the efficacy of adoptively transferred cells in preclinical models. There are still some unaddressed aspects of CAR T cell biology and functionality, such as understanding how epigenetic reprogramming and gene regulation enhance CAR T cell antitumor function.4–6 The central dogma of molecular biology has come under scrutiny by the emerging field of epigenetics.

    • Anti-tumor effects of vascular endothelial growth factor/vascular endothelial growth factor receptor binding domain-modified chimeric antigen receptor T cells

      2021, Cytotherapy
      Citation Excerpt :

      Combination therapies of CAR T cells with various small molecules and monoclonal antibodies would contribute to circumvent tumor escape and increase anti-tumor activity to achieve complete tumor eradication [44].

    View all citing articles on Scopus
    View full text