Research review paper
CAR-T immunotherapies: Biotechnological strategies to improve safety, efficacy and clinical outcome through CAR engineering

https://doi.org/10.1016/j.biotechadv.2019.06.010Get rights and content

Abstract

T cells engineered to express a chimeric antigen receptor (CAR) have re-shaped the way hematological malignancies are treated. Despite the overwhelming early clinical success, CAR-T therapies are associated with severe side-effects, disease relapse and often exhibit limited efficacy. In this Review article we summarize the most recent biotechnological advances that have been developed to enhance the efficacy and specificity of CAR-T therapies, as well as to address the key challenges associated with them. We place particular emphasis on the most recent clinical data that indicate which CAR-T populations are the most relevant to clinical success, and indicate how the molecular structure of the CAR receptor can affect clinical outcome. Finally, we outline what we believe is the next generation of immunotherapies.

Introduction

Two autologous chimeric antigen receptor (CAR)-T cell therapies (Kymriah and Yescarta) were recently licensed by American and European agencies (FDA and EMA respectively). CAR-T cell therapies are a type of cancer immunotherapy in which a patient’s T cells are genetically engineered in the laboratory so they can attack cancer cells. Both licensed products target CD19, an antigen expressed on B cells and leukemic cells. Kymriah (tisagenlecleucel) brought to market by Novartis and Yescarta (axicabtagene ciloleucel) brought to market by Kite Pharma/ Gilead, are indicated for the treatment of pediatric and relapse/refractory (R/R) B cell acute lymphoblastic leukemia (B-ALL) and certain lymphoma subtypes and adult refractory diffuse large B cell lymphoma (R/DLBCL) respectively. Remarkably, the overall response rate in the short term was 83% based on a single dose of Kymriah with patients entering into remission within 3 months of being treated (Grupp, 2018; Maude et al., 2018; Maude et al., 2014) (ELIANA, NCT02435849) (Table 2), extending to 76% and 70% at 12 months and 18 months respectively (Grupp, 2018). Similarly, the recipients of Yescarta had an overall response rate of 71% within 3 months (Locke et al., 2018; Locke et al., 2015; Long et al., 2015; Neelapu et al., 2017) (ZUMA, NCT02348216) and 39% of patients remained in remission for 27 months (Locke et al., 2018; Neelapu, 2018).

Although CAR-T therapies have a remarkable life-saving potential they are nevertheless associated with severe toxicities (most notably cytokine release syndrome and neurotoxicities), high cost (> £200,000) and limited efficacy on cancers other than blood malignancies. Furthermore, despite the high short-term response rates a proportion of patients relapse after CAR-T treatment. As a result, further understanding of T and CAR-T cell biology is required in order to improve these immunotherapies.

The immune system is comprised of the innate and adaptive systems. While the innate system is the first line of defense and is poised to act rapidly, the adaptive system displays immunological memory and contains cells such as T and B cells that are highly specific for any pathogenic threat. T cells are defined by the expression of the T cell receptor (TCR) which interacts with the Major Histocompatibility Complex (MHC) (also termed human leukocyte-associated [HLA] antigens), found on the surface of antigen presenting cells (APCs) such as dendritic cells (DC). This interaction is necessary for the recognition of the pathogenic threat by the T cell and its subsequent activation (Fig. 1). The immunogenic response elicited by the T cell is dependent on the type of MHC that is presented by the APC (class I MHC or class II MHC), which subsequently defines whether the T cell will differentiate into one of the two major T cell subsets, CD8+ (T cytotoxic) or CD4+ (T helper). While most T cells disappear after the threat has been eliminated, others will survive and form memory cells that can survive for years. Memory T cells are classified according to the expression of certain surface markers (Table 1) (Gattinoni et al., 2011; Gattinoni et al., 2009; Gebhardt et al., 2009; Hofmann and Pircher, 2011; Lugli et al., 2013; Masopust et al., 2010; Sallusto et al., 1999).

This review outlines the challenges associated with CAR-T immunotherapies and focuses on the most recent biotechnological and genomic engineering advancements that have been developed in order to address such challenges. Furthermore the review summarizes the recent CAR-T clinical data that provide an indication as to which subsets of CAR-T cells have the most potential to expand within the patient and therefore provide sustainable remissions. Finally the review discusses alternative CAR immunotherapies by utilizing other cells of the immune system. The review is therefore intended for individuals with a biochemical engineering, biotechnology or similar background who wish to further understand CAR-T immunotherapies from a biological or biotechnological perspective.

Section snippets

CAR design

Genetically engineered T cells ectopically expressing CAR are referred to as CAR-T cells. Unlike TCRs, CARs enable highly-specific targeting of antigen in an MHC-independent manner. CARs comprise of extracellular, hinge/transmembrane and intracellular domains (Fig. 2). The CAR design has evolved over the years in order to enhance safety and efficacy (Fig. 2). First generation CARs were reported by Kuwana and Gross and later by Eshhar et al and only included the CD3ζ domain (Kuwana et al., 1987;

Alternative CAR immunotherapies

As the field of immunotherapies continues to develop, alternative avenues are currently being explored. Notable examples include a distinct and less common type of T cells, gamma delta T cells (γδ T). Unlike conventional T cells that are currently used for CAR-T therapies and express αβ TCR, γδ T cells express γδ TCR. Furthermore, while conventional T cells rely on specific antigens for target recognition, γδ T cells recognize “stress-antigens” which are indicative of malignant transformation

Concluding remarks and future outlook

In the past year, the CD19 CAR-T products Kymriah and Yescarta have gained approval by the European Medicines Agency (EMA) for their use in the European Union following their approval by the FDA. While Kymriah was granted approval by the UK’s National Institute for Healthcare and Excellence (NICE) for its use to treat children and adults up to 25 years suffering from B-ALL, it was turned down for its use in adults suffering from DLBCL, a disease affecting more than 4,800 people in the UK (//lymphoma-action.org.uk

Acknowledgements

The authors would like to acknowledge the EPSRC for funding this work through the “New Industrial Systems: Optimising Me Manufacturing Systems”, Grant Number EP/R022534/1 and “Future Targeted Healthcare Manufacturing Hub”, Grant Number EP/P006485/1.

References (142)

  • X. Han et al.

    Masked chimeric antigen receptor for tumor-specific activation

    Mol. Ther. J. Am. Soc. Gene Ther.

    (2017)
  • B. Hu et al.

    Augmentation of antitumor immunity by human and mouse CAR T cells secreting IL-18

    Cell Rep.

    (2017)
  • S. Jaiswal et al.

    CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis

    Cell

    (2009)
  • O.U. Kawalekar et al.

    Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells

    Immunity

    (2016)
  • Y. Kuwana et al.

    Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions

    Biochem. Biophys. Res. Commun.

    (1987)
  • C.H. Lamers et al.

    Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity

    Mol. Ther. J. Am. Soc. Gene Ther.

    (2013)
  • H.-W. Lee et al.

    Recruitment of monocytes/macrophages in different tumor microenvironments

    Biochim. Biophys. Acta

    (2013)
  • D.W. Lee et al.

    T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial

    Lancet Lond. Engl.

    (2015)
  • H.G. Ljunggren et al.

    In search of the “missing self”: MHC molecules and NK cell recognition

    Immunol. Today

    (1990)
  • F.L. Locke et al.

    Phase 1 clinical results of the ZUMA-1 (KTE-C19-101) study: a phase 1-2 multi-center study evaluating the safety and efficacy of anti-CD19 CAR T Cells (KTE-C19) in subjects with refractory aggressive non-hodgkin lymphoma (NHL)

    Blood

    (2015)
  • K.-J. Malmberg et al.

    Natural killer cell-mediated immunosurveillance of human cancer

    Semin. Immunol.

    (2017)
  • J.C. Markley et al.

    IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient mice

    Blood

    (2010)
  • 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. J. Am. Soc. Gene Ther.

    (2009)
  • R.A. Morgan et al.

    Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2

    Mol. Ther. J. Am. Soc. Gene Ther.

    (2010)
  • D. Abate-Daga 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)
  • G.L. Beatty et al.

    Chimeric antigen receptor T cells are vulnerable to immunosuppressive mechanisms present within the tumor microenvironment

    Oncoimmunology

    (2014)
  • J.C. Becker et al.

    Immune-suppressive properties of the tumor microenvironment

    Cancer Immunol. Immunother.

    (2013)
  • R.J. Brentjens et al.

    CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia

    Sci. Transl. Med.

    (2013)
  • R.J. Buckanovich et al.

    Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy

    Nat. Med.

    (2008)
  • C. Carpenito et al.

    Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • C.T. Charlesworth et al.

    Identification of Pre-existing Adaptive Immunity to Cas9 Proteins in Humans

    (2018)
  • D. Chinnasamy et al.

    Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2012)
  • J.H. Cho et al.

    Universal chimeric antigen receptors for multiplexed and logical control of T cell responses

    Cell

    (2018)
  • S.E. Church et al.

    Tumor-specific CD4+ T cells maintain effector and memory tumor-specific CD8+ T cells

    Eur. J. Immunol.

    (2014)
  • R. Clarke

    Pluripotent cell-derived off-the-shelf TCR-less CAR-targeted cytotoxic T cell therapeutic for the allogeneic treatment of B cell MALIGNANCIES

  • J.A. Craddock et al.

    Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b

    J. Immunother.

    (2010)
  • J.G. Crompton et al.

    Uncoupling T-cell expansion from effector differentiation in cell-based immunotherapy

    Immunol. Rev.

    (2014)
  • M.L. Davila et al.

    Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia

    Sci. Transl. Med.

    (2014)
  • Z. Eshhar et al.

    Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors

    Proc. Natl. Acad. Sci. U. S. A.

    (1993)
  • A.G. Evans et al.

    Evolution to plasmablastic lymphoma evades CD19-directed chimeric antigen receptor T cells

    Br. J. Haematol.

    (2015)
  • V.D. Fedorov et al.

    PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses

    Sci. Transl. Med.

    (2013)
  • H.M. Finney et al.

    Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product

    J. Immunol. Baltim. Md

    (1998)
  • H.M. Finney et al.

    Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain

    J. Immunol. Baltim. Md

    (2004)
  • J.A. Fraietta et al.

    Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia

    Nat. Med.

    (2018)
  • J.A. Fraietta et al.

    Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells

    Nature

    (2018)
  • T.J. Fry et al.

    CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy

    Nat. Med.

    (2018)
  • L. Gattinoni et al.

    Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells

    J. Clin. Invest.

    (2005)
  • L. Gattinoni et al.

    Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells

    Nat. Med.

    (2009)
  • L. Gattinoni et al.

    A human memory T cell subset with stem cell-like properties

    Nat. Med.

    (2011)
  • T. Gebhardt et al.

    Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus

    Nat. Immunol.

    (2009)
  • Cited by (14)

    • An impedimetric immunosensor for the selective detection of CD34<sup>+</sup> T-cells in human serum

      2022, Sensors and Actuators B: Chemical
      Citation Excerpt :

      Recently, Chimeric Antigen Receptor T (CAR-T) cell-based immunotherapy, which combines the specificity of antibodies to their antigens with the cytotoxic potential of T-cells, has shown great potential as an effective strategy for personalized cancer treatment [3]. This therapy, approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA), has in particular shown clinical efficacy for acute lymphoblastic leukaemia, chronic lymphocytic leukaemia, and non-Hodgkin’s lymphoma [4–6]. CAR-T cells are manufactured by genetically engineering patients’ native T-cells ex vivo, to specifically recognize tumour antigens [7].

    • Objectives, benefits and challenges of bioreactor systems for the clinical-scale expansion of T lymphocyte cells

      2021, Biotechnology Advances
      Citation Excerpt :

      With such approaches, normal T cells from patients suffering from other cancers are isolated through apheresis and transduced to generate TCR or CAR T cells (Barrett et al., 2015) (Fig. 3). Details on these methods and their development have been previously reported in review papers (Barrett et al., 2015; Dai et al., 2018, 2019; Field and Qasim, 2015; Mellman et al., 2011; Panagopoulou and Rafiq, 2019; Piscopo et al., 2018). An activation procedure has to be performed to allow for transduction and proliferation of the manufactured T cells.

    • Anti-tumour effect of the fourth-generation chimeric antigen receptor T cells targeting CD133 against cholangiocarcinoma cells

      2020, International Immunopharmacology
      Citation Excerpt :

      Further evaluations of anti-CD133-CAR4 T cell reactivity in normal tissues, and its optimal therapeutic window in a clinical setting are required. To ensure safety, the anti-CD133-CAR4 T cells may be modified to harbour suicide genes [e.g., herpes simplex virus thymidine kinase (HSVtk), inducible caspase 9 (iCasp9), or truncated epithelial growth factor receptor (tEGFR)], which would allow elimination of CAR T cells, if required [12,58]. Future studies should investigate combination of anti-CD133-CAR4 T cells and patient-preconditioning with other standard treatments, including chemotherapy using gemcitabine or cisplatin [59], immune checkpoint inhibitors [60], or in situ secretion of bispecific T cell engager [61,62] to overcome different obstacles to cancer eradication.

    View all citing articles on Scopus
    View full text