Elsevier

Blood Reviews

Volume 41, May 2020, 100645
Blood Reviews

Review
Autoimmunity as a target for chimeric immune receptor therapy: A new vision to therapeutic potential

https://doi.org/10.1016/j.blre.2019.100645Get rights and content

Abstract

Chimeric immune receptors (CIRs) are functionally pleiotropic because they are artificially expressed on diverse cell types, which gives specificity to their function to anergize, kill, or protect cognate target cells. CIRs consist of chimeric antigen receptors (CARs) and B-cell antibody receptor (BAR) or chimeric autoantibody receptors (CAARs). Approval of CAR-T cell therapy by the Food and Drug Administration (FDA) has encouraged investigators to search for autoimmune therapies that are CIR-based. Both T effector cells, particularly CD8+, and T CD4+ regulatory cells (Tregs) can be engineered through CIR expression. Recently, natural killer cells have been included to increase efficiency. Unwanted antibody producer B cells are effectively prevented by CAAR-T cells, B-cell antibody receptor (BAR)-T CD8+, and BAR-Treg, which represents an advantage in antibody-mediated diseases such as pemphigus vulgaris (PV) and hemophilia A. Although CAAR and BAR-T cells may have curative benefits for autoantibody-mediated immune diseases, verification of long-term efficacy and safety are a priority before clinical use. Effective CIR-T cell therapy largely depends on the reliability and stability of the receptor. Based on CIR functionality, factors that explicitly determine effectiveness of the treatment should be considered. These factors include antigen/autoantibody specificity, single chain variable fragment (scFv) affinity, and autoantibody masking. Herein, we review the current evidence of CIR therapy with a focus on their therapeutic potential for autoimmune diseases and their challenges.

Introduction

Adaptive T cell therapy is a new and rapidly developing area in cell therapy in which the adoptive transfer of genetically reprogrammed T cells, such as TCR transgenic cells and chimeric immune receptors (CIRs), has received more attention because they form recombinant or new receptors that can improve specific-antigen recognition [1].

TCR transgenic structures are designed to enhance affinity of the TCR for peptide-MHC. In TCR engineering T cell therapy, antigen specificity is determined by the new TCR heterodimer-mediated signaling pathway. However, during TCR engineering, more and sustainable responses are achieved when retroviral or lentiviral vectors are used to molecularly clone TCR-α and -β subunits from highly reactive T cells [1,2]. TCR engineering is a tool that can be used to augment T cells. TCR engineered T effector cells (Teff) are designed to target disruption, whereas TCR engineered regulatory T (Treg) cells can anergize/kill and protect target cells [3].

Although TCR engineering T cell therapy is a promising strategy to induce defined antigen-specific immune responses, limitations to their use include MHC-polymorphism, peptide antigens, unpredictable mixed TCR dimers, and TCR cross-reactivity against related or unrelated antigens [2].

CIR-T cells are of tremendous interest in T cell therapies for hematopoietic and solid malignancies, and immune-mediated diseases. Advantages of CIR-T cells include increased specificity and sensitivity to target cells, their non-MHC dependent manner, and extended safety [4].

The privileged activation of T cells following stimulation of CIR make them promising in an MHC-independent manner, which gives them a universal therapeutic use and superiority to TCR-based T cell therapies [4,5].

Generally, CIRs have two functional domains and a transmembrane region that fit the task to accomplish and are involved in signal transduction. The extracellular domain consists of a chopped fragment of a monoclonal antibody called a single chain variable fragment (scFv) in CAR or a specific antigen in CAAR, which can be a bio-therapeutic agent in a BAR system [6]. These domains bind to target antigens that are relatively specific for cells, tissues, and organs and to autoantibodies in CARs and BARs. Fig. 1 provides a summary of different CIR-T cells, including CAR-Treg and BAR-Teffs that protect normal cells. At the other end of the CIRs, signal transduction is initiated through the TCR domain. Thus, it becomes clear that CD3ζ plays a prominent role in the CIR system and other members can be exchanged for improvement without attenuating its function [7].

The lack of tissue-specific localization and antigen-specific detection substantially influences therapies for autoimmune diseases. These can be affected by CIR-T cells, resulting in eventual disruption or protection of targeted cells in various autoimmune diseases such as multiple sclerosis (MS), type 1 diabetes (T1D), colitis, rheumatoid arthritis (RA), and pemphigus vulgaris (PV) [8]. In terms of antigen-specific T cells for autoimmune diseases, the CIR system can be categorized into BAR-T cells and CAR-Tregs that exogenously express CIR [9,10].

While CAR-T CD8+ professionally kill the tumor cells, the purpose of CIR-T cell therapy for autoimmune diseases is to provide direct protection of normal cells by CAR-Tregs and/or indirect protection by BAR-T cells (Fig. 2) [6,[10], [11], [12]].

Recently, CIR engineered T cells have been introduced as a protective factor against antibody-mediated responses in bio-therapeutics such as FVIII [12]. Zhang et al. conducted a CIR-based intervention to suppress antibody-mediated responses to factor VIII (FVIII) through BAR-Treg-mediated FVIII-specific B-cell suppression [12]. The FVIII-specific antibody response decreased through two distinct mechanisms, FVIII-specific B cells that were directly tolerized to the suppressive activity of FVIII-specific BAR-Tregs by currently unknown mechanisms and via indirect suppression of B cell function through suppression of T effector cells, which are essential for B cell activation [12]. Another aspect of B cell suppression was reported by Parvathaneni and Scott [6]. They used FVIII-specific BAR-CD8+ T cells to directly eliminate FVIII-specific B cells [6]. However, these evidences led to the conclusion that remarkable suppression of unwanted antibody production was due to BAR-Treg and BAR-T CD8+ therapy [6,12]. Clearly, it would be useful for future prophylactic treatment of patients who receive bio-therapeutic drugs such as FVIII. These results have suggested that B cell-mediated antibody responses can be controlled by BAR T cell system like dominant responses that occurred during antibody-mediated autoimmune diseases.

Although there are numerous clinical reports of CIR-T cell therapy in hematopoietic and solid tumor malignancies [[13], [14], [15], [16]], only preclinical studies have assessed the effect of CIR-T cells on a few autoimmune diseases. One clinical trial evaluated the induction of immune tolerance by polyclonal Tregs exclusively in new onset T1D, it and other ongoing trials listed in Table 1 [17].

Section snippets

Chimeric antigen receptor (CAR) characteristics and design

Tumor-infiltrating lymphocyte (TILs) therapy faces several challenges in processing and is potentially time-consuming; however, new immunotherapy approaches, such as genetically manipulated T cells, are making progress to overcome these challenges [18,19]. In 1993, CAR produced by genetically engineered T cells was first proposed by Eshhar et al. [20]. They used scFv from a target antibody and combined it with a cytoplasmic phosphorylation domain of the CD3ζ to direct engineered T cells toward

The therapeutic potential of chimeric immune receptors (CIRs) to treat autoimmune diseases

In autoimmune disease therapy, CIRs precisely target either humoral or cellular immunity, and indirectly affect the complement system. The CAR-based autoimmune therapy is vary widely from cancer therapy with regards to CAR design and antigen targeting [39]. The developed CIRs system attempts to control unwanted immune responses in autoimmune diseases; nonetheless, it is still in its infancy stage and additional research is necessary to achieve a safe, more efficient therapy [40,41]. In a few

Antigen-specific tolerance via CAR-Tregs for autoimmune diseases

Sakaguchi et al. reported the first evidence for reestablishment of peripheral tolerance by Tregs, a subset of CD4+ T cells [49]. Tregs can be divided into two major categories based on developmental pathways, natural Tregs (nTregs) that arise from the thymus and induced Tregs (iTregs) that are formed in the peripheral immune system. Tregs are identified by loss of CD127 in addition to elevated expression of CD25 and intracellular transcription factor forkhead box P3 (Foxp3). The

Antigen-specific suppression via CIR-killer cells for treatment of autoimmune diseases

Autoantibody production by B cells is often an autoimmune reaction for the onset of antibody-mediated diseases. Monoclonal antibodies against the B-cell surface antigen, CD20, cause selective transient depletion of B cells, and lead to general immunosuppression followed by hypogammaglobulinemia [74]. The targeted depletion of specific subsets of B cells by CAAR-T cells is an effective alternative strategy to antibody therapy that reduces the risks of general B cell depletion by antibodies such

CIR-T cell therapy challenges

While there are some similarities in generation and function, differences do exist between tumor targeting and normal tissue targeting in CIR cell-based immunotherapy. The use of CIR-T cells for protection and/or killing of target cells in autoimmune diseases provides a new therapeutic window that has numerous challenges [40,83].

Future considerations

Overall, CAR-Treg for autoimmunity has undergone more rapid development than CAR- and CAAR-T cells. CAR-Treg are becoming a popular issue in immunology because of their potential for restoration of self-tolerance, one of the most important properties of the immune system that is lost in autoimmunity. The developed CIR system especially responds to the stable conformation of cell-surface expressed antigens, which should be specific to target cells, tissues, and organs. Investigators have tackled

Conclusion

CAR-Treg and CAAR-Teff therapies are two major T cell engineering strategies that have been proposed for immunotherapy of autoimmune disorders. Advances in CIR modified T cells for cancer immunotherapy have paved the way for application of the same principles to treat autoimmune diseases. Antigen recognition with CIRs and subsequent signal transduction follow the same principles in both Teffs and Tregs, although the CIR domains are different. Clinical grade manufacturing of cancer-specific

Practice points

  • Due to CIR specificity, CIR-T cell therapy could be employed to redirect T cells toward autoreactive T and B cells in autoimmune disorders.

  • CIR-cell immunotherapy is a personalized therapy with the potential for a long-term disease-free period for autoimmune patients. It can be presented in the context of MHC-independent therapy without general immunosuppression.

  • CAR-Treg therapy is a new insight into cell-mediated antigen-specific immune tolerance induction; however, it has not been practically

Research agenda

  • Characterization of intracellular domains to prolong persistence and high activation.

  • Characterization of the off-targeting and cytokine-associated toxicity associated with CIR-T cells in different autoimmune diseases.

  • Determination of the specific antigen, localization, and long-term persistence in cell-mediated autoimmune diseases.

  • Development of assays to obtain Treg expansion and de novo generation.

  • Development of assays for high transduction efficiency.

Declaration of Competing Interests

The authors have no conflicts of interest.

Author contributions

M.H H wrote the original draft and created the figures and tables. M.H H and E H-S participated in the design. E H-S, H B and B N reviewed and edited the final manuscript. All authors have approved the final manuscript.

Funding

Supported by grants from Stem Cell Biology and Technology Department of Royan Institute (95000278) Iran, Tehran University of Medical Sciences, Iran, and Royan Stem Cell Technology Company (96030204 RFCT), Iran.

Acknowledgments

The authors gratefully acknowledge the Stem Cell Biology and Technology Department of Royan Institute, Tehran University of Medical Sciences, and Royan Stem Cell Technology Company for financial support.

References (118)

  • C Sommer et al.

    Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma

    Mol Ther

    (2019)
  • X Han et al.

    Adnectin-based design of chimeric antigen receptor for T cell engineering

    Mol Ther

    (2017)
  • Z Grada et al.

    TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy

    Mol Ther-Nucl Acids

    (2013)
  • E Elinav et al.

    Amelioration of colitis by genetically engineered murine regulatory T cells redirected by antigen-specific chimeric receptor

    Gastroenterology.

    (2009)
  • A Lucchese

    Streptococcus mutans antigen I/II and autoimmunity in cardiovascular diseases

    Autoimmun Rev

    (2017)
  • B-S Kim et al.

    Generation of RORγt+ antigen-specific T Regulatory 17 cells from Foxp3+ precursors in autoimmunity

    Cell Rep

    (2017)
  • AK Shakya et al.

    Antigen-specific Tolerization and targeted delivery as therapeutic strategies for autoimmune diseases

    Trends Biotechnol

    (2018)
  • K Earle et al.

    In vitro expanded human CD4+ CD25+ regulatory T cells suppress effector T cell proliferation

    Clin Immunol

    (2005)
  • A Ma et al.

    Dysfunction of IL-10-producing type 1 regulatory T cells and CD4+ CD25+ regulatory T cells in a mimic model of human multiple sclerosis in Cynomolgus monkeys

    Int Immunopharmacol

    (2009)
  • T Magg et al.

    IPEX due to an exon 7 skipping FOXP3 mutation with autoimmune diabetes mellitus cured by selective TReg cell engraftment

    Clin Immunol

    (2018)
  • S McEwen et al.

    Cell therapy in transplantation

  • N Marek-Trzonkowska et al.

    Therapy of type 1 diabetes with CD4+ CD25highCD127-regulatory T cells prolongs survival of pancreatic islets—results of one year follow-up

    Clin Immunol

    (2014)
  • E Elinav et al.

    Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice

    Gastroentrology.

    (2008)
  • D Blat et al.

    Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells

    Mol Ther

    (2014)
  • SE Gitelman et al.

    Regulatory T cell therapy for type 1 diabetes: may the force be with you

    J Autoimmun

    (2016)
  • G Boleto et al.

    Predictors of hypogammaglobulinemia during rituximab maintenance therapy in rheumatoid arthritis: a 12-year longitudinal multi-center study

  • H Meng et al.

    La/SSB chimeric autoantibody receptor modified NK92MI cells for targeted therapy of autoimmune disease

    Clin Immunol

    (2018)
  • JA McIntyre et al.

    Autoantibodies unmasked by redox reactions

    J Autoimmun

    (2005)
  • JN Brudno et al.

    Toxicities of chimeric antigen receptor T cells: recognition and management

    Blood.

    (2016)
  • H Jethwa et al.

    Use of gene-modified regulatory T-cells to control autoimmune and alloimmune pathology: is now the right time?

    Clin Immunol

    (2014)
  • D Maimone et al.

    IL-6 detection in multiple sclerosis brain

    J Neurol Sci

    (1997)
  • K Ichiyama et al.

    Foxp3 inhibits RORγt-mediated IL-17A mRNA transcription through direct interaction with RORγt

    J Biol Chem

    (2008)
  • SJ Schuster et al.

    Chimeric antigen receptor T cells in refractory B-cell lymphomas

    N Engl J Med

    (2017)
  • TT Spear et al.

    Understanding TCR affinity, antigen specificity, and cross-reactivity to improve TCR gene-modified T cells for cancer immunotherapy

    Cancer Immunol Immunother

    (2019)
  • YC Kim et al.

    Engineered antigen-specific human regulatory T cells: immunosuppression of FVIII-specific T-and B-cell responses

    Blood.

    (2015)
  • DM Barrett et al.

    Chimeric antigen receptor–and TCR-modified T cells enter main street and wall street

    J Immunol

    (2015)
  • CR Maldini et al.

    CAR T cells for infection, autoimmunity and allotransplantation

    Nat Rev Immunol

    (2018)
  • S Patel et al.

    Impact of chimeric immune receptor extracellular protein domains on T cell function

    Gene Ther

    (1999)
  • A-H Zhang et al.

    Targeting antigen-specific B cells using antigen-expressing transduced regulatory T cells

    J Immunol

    (2018)
  • A-H Zhang et al.

    Targeting FVIII-specific B cells using BAR-transduced regulatory T cells

    Blood.

    (2016)
  • D Littman et al.

    Cancer immunotherapy with chimeric antigen receptor (CAR) T cells

    J Onco-Nephrol

    (2017)
  • EJ Grigor et al.

    Efficacy and safety of chimeric antigen receptor T-cell (CAR-T) therapy in patients with haematological and solid malignancies: protocol for a systematic review and meta-analysis

    BMJ Open

    (2017)
  • HR Mirzaei et al.

    Chimeric antigen receptors t cell therapy in solid tumor: challenges and clinical applications

    Front Immunol

    (2017)
  • JA Bluestone et al.

    Type 1 diabetes immunotherapy using polyclonal regulatory T cells

    Sci Transl Med

    (2015)
  • SA Rosenberg et al.

    Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy

    Clin Cancer Res

    (2011)
  • 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

    P Natl Acad Sci USA

    (1993)
  • YJ Xie et al.

    Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice

    P Natl Acad Sci USA

    (2019)
  • SJ Parsons et al.

    Src family kinases, key regulators of signal transduction

    Oncogene.

    (2004)
  • AK Chakraborty et al.

    Insights into the initiation of TCR signaling

    Nat Immunol

    (2014)
  • B Savoldo et al.

    CD28 costimulation improves expansion and persistence of chimeric antigen receptor–modified T cells in lymphoma patients

    J Clin Invest

    (2011)
  • Cited by (18)

    • Human stem cell-derived thymic epithelial cells enhance human T-cell development in a xenogeneic thymus

      2022, Journal of Allergy and Clinical Immunology
      Citation Excerpt :

      The TCR sequencing data were retrieved from Adaptive Biotechnologies’ ImmunoSEQ software. Computational and statistical analysis was performed using R software (R Foundation, Vienna, Austria), as we previously described.17,18 Statistical analysis and comparisons were performed with GraphPad Prism 7.0 (GraphPad Software, San Diego, Calif).

    • Clinical and diagnostic potential of regulatory T cell markers: From bench to bedside

      2022, Transplant Immunology
      Citation Excerpt :

      The clinical success of adoptive T cell immunotherapy has opened up new opportunities for treating autoimmune disorders and obtaining better outcomes after allograft transplantation, and this is because Treg cells are supposed to play a central role in immune tolerance induction [5–8]. Treg therapy has emerged over the past several years as a promising method for tolerance induction, and several clinical trials are finished, currently recruiting patients, or now open for enrollment [4,9]. Treg surface markers are important for antibody-based detection and isolation of these cells.

    • Intelligent cell-based therapies for cancer and autoimmune disorders

      2020, Current Opinion in Biotechnology
      Citation Excerpt :

      The aforementioned strategies involve the engineering of effector T cells that may potentially lead to serious complications in cases with off-target activation [49]. To circumvent this caveat, Raffin et al. proposed to harness the immunosuppressive power of regulatory T cells (Tregs) [49,50]. Tregs exert bystander suppression and enable control of autoimmunity by trafficking into sites of inflammation without specific cell depletion.

    View all citing articles on Scopus
    View full text