Elsevier

Seminars in Immunology

Volume 52, February 2021, 101473
Seminars in Immunology

Review
Checkpoint blockade toxicities: Insights into autoimmunity and treatment

https://doi.org/10.1016/j.smim.2021.101473Get rights and content

Abstract

Checkpoint blockade has transformed not only the way cancers are treated, but also highlighted the importance of mounting a proper immune response against tumors. Despite advances in the field of immunotherapy, many patients develop a range of inflammatory toxicities that limit the efficacy of these therapies. These toxicities range from barrier site injury, such as colitis, to endocrine organ dysfunction, such as diabetes. In order to properly treat patients with cancer and avoid checkpoint blockade induced toxicities, we must gain a deeper understanding of the underlying mechanisms generating these adverse events. Cytotoxic and tissue-resident T cells likely play an important role in mediating some toxicities, though high levels of cytokines and the generation of auto-antibodies in other toxicities demonstrates these mechanisms are not all shared. Certain risk factors for specific toxicities may be able to predict who might benefit most from alternative therapies given the risk-benefit associated with checkpoint blockade. As the targets of checkpoint inhibitors have important functions in the prevention of autoimmunity, insights into risk factors and causes of toxicities will further our knowledge of fundamental immunology and enable the development of novel therapeutics.

Introduction

Monoclonal antibody blockade of the immune regulatory “checkpoint” receptors cytotoxic T lymphocyte antigen (CTLA)-4, Programmed Death (PD)-1 and its ligand PD-L1 has revolutionized the treatment of diverse cancers, leading to durable responses in a subset of patients with advanced malignancies [1,2]. The clinical success of checkpoint blockade has underscored the importance of endogenous immune responses in shaping cancer growth, and revealed a critical role for reactivation of effector memory CD8 + T cells in mediating this response [3]. However, alongside the benefits of checkpoint blockade has come a wide range of inflammatory toxicities. These toxicities can affect any organ in the body and frequently cause morbidity and delayed or discontinued immunotherapy use; in rare cases, these toxicities can even be fatal [4,5]. Inflammation from checkpoint blockade most commonly affects the barrier tissues, including the skin, gastrointestinal tract and liver, and the respiratory epithelium [5]. Endocrine organs and joints are also common targets, similar to many spontaneous autoimmune diseases [6,7]. Inflammation in the heart and nervous system is rare, affecting only a few percent of people on checkpoint blockade, but both are important cases of fatal toxicity [4]. CTLA-4 blockade is considerably more likely to cause inflammatory side effects than either PD-1 or PD-L1 blockade, and for CTLA-4 inhibitors, the frequency and severity of this inflammation is dose dependent [5,[8], [9], [10]]. When CTLA-4 and PD-1 blockade are combined to treat melanoma, nearly all patients develop some inflammatory side effects, and treatment-limiting toxicities occur in over half of patients [11].

In addition to the impact these inflammatory side effects have on clinical care, these toxicities are also an important limitation to developing novel treatments. Checkpoint inhibitor toxicities are more common with combination immunotherapies, and also occur more frequently when combined with other treatment modalities such as targeted agents and chemotherapy [[11], [12], [13], [14], [15]]. An expansive array of immunotherapies targeting other pathways are in clinical development, including antibodies targeting other co-inhibitory receptors (e.g. TIM3, LAG3 and TIGIT), agonistic antibodies against co-stimulatory receptors (e.g. OX40, CD137, CD40, GITR), as well as a variety of other immune modulating agents acting on multiple distinct pathways [16]. Many of these agents have been limited by adverse events, or dosage has been reduced to avoid them. This is particularly true for novel combination treatments, which are also more likely to generate a clinical response.

Section snippets

Checkpoint blockade toxicities as a model for understanding immune regulation in humans

The inflammatory toxicities of checkpoint blockade offer important insights into the roles of CTLA-4, PD-1 and PD-L1 in immune regulation. Because receptor inhibition causes toxicity, understanding the mechanism driving these toxicities will deepen our understanding of immune regulation by CTLA-4, PD-1, and PD-L1.

Risk factors for checkpoint blockade toxicities

Establishing risk factors to predict the development of specific checkpoint blockade toxicities is of considerable clinical importance. Many of these toxicities are treatment-limiting or can be life-threatening [4]. Predicting who might develop them could allow for testing of prophylactic therapy, as has been done unsuccessfully for checkpoint colitis [38], and could also alter decisions about the risk-benefit balance prior to treatment. Improving our prediction of risk and benefit is of

Balancing antitumor immunity and immune suppression to treat checkpoint blockade toxicities

Most checkpoint blockade toxicities respond to treatment with systemic glucocorticoids [49,[52], [53], [54]]; however, these drugs are broadly immune suppressive, and can induce apoptosis in activated T cells, suggesting a potentially deleterious effect on antitumor responses. Endogenous glucocorticoids have also recently been implicated in development of dysfunctional T cells [55]. Although definitive data from humans is lacking, increasing data suggest the potential for an inhibitory effect

Concluding thoughts

The inflammatory toxicities of checkpoint blockade are becoming increasingly more important, limiting the treatment efficacy of approved immunotherapies while also serving as a barrier to novel combinations. Efforts to study the mechanisms of these toxicities not only have the potential to lead to substantial clinical benefit by identifying novel treatment approaches but may also teach important biology. These toxicities represent the in vivo consequence of loss of CTLA-4, PD-1 and PD-L1

Declaration of Competing Interest

M.D. is a consultant for Tillotts Pharma, Partner Therapeutics, Genentech-Roche, and Moderna and receives research funding from Novartis and Eli Lilly, and is on the Scientific Advisory Board for Neoleukin Therapeutics.

Acknowledgments

This work was supported by a Team Award from the American Cancer Society <GN1>M</GN1>RAT-18-113-01 and the Melanoma Research Alliance597698 (M.D.), and a National Institutes of Health Mentored Clinical Scientist Development Award1K08DK114563-01 (M.D.).

References (83)

  • A. Ribas et al.

    Cancer immunotherapy using checkpoint blockade

    Science

    (2018)
  • S.H. Baumeister et al.

    Coinhibitory pathways in immunotherapy for cancer

    Annu. Rev. Immunol.

    (2016)
  • M. Sade-Feldman

    Defining T cell states associated with response to checkpoint immunotherapy in melanoma

    Cell

    (2018)
  • D.Y. Wang

    Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis

    JAMA Oncol.

    (2018)
  • M. Dougan

    Checkpoint blockade toxicity and immune homeostasis in the gastrointestinal tract

    Front. Immunol.

    (2017)
  • L.S. Chang

    Endocrine toxicity of Cancer immunotherapy targeting immune checkpoints

    Endocr. Rev.

    (2019)
  • L.C. Cappelli et al.

    Rheumatic and musculoskeletal immune-related adverse events due to immune checkpoint inhibitors: a systematic review of the literature

    Arthritis Care Res. (Hoboken)

    (2017)
  • F.S. Hodi

    Improved survival with ipilimumab in patients with metastatic melanoma

    N. Engl. J. Med.

    (2010)
  • A.M. Eggermont

    Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy

    N. Engl. J. Med.

    (2016)
  • C. Robert

    Pembrolizumab versus ipilimumab in advanced melanoma

    N. Engl. J. Med.

    (2015)
  • J. Larkin

    Combined nivolumab and ipilimumab or monotherapy in untreated melanoma

    N. Engl. J. Med.

    (2015)
  • L. Paz-Ares

    Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer

    N. Engl. J. Med.

    (2018)
  • B.I. Rini

    Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma

    N. Engl. J. Med.

    (2019)
  • P. Schmid

    Atezolizumab and nab-paclitaxel in advanced triple-negative breast Cancer

    N. Engl. J. Med.

    (2018)
  • M.D. Hellmann

    Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden

    N. Engl. J. Med.

    (2018)
  • M. Dougan et al.

    Cancer immunotherapy: beyond checkpoint blockade

    Annu. Rev. Cancer Biol.

    (2019)
  • P.A. van der Merwe et al.

    CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics

    J. Exp. Med.

    (1997)
  • J.C. Schwartz et al.

    Structural basis for co-stimulation by the human CTLA-4/B7-2 complex

    Nature

    (2001)
  • O.S. Qureshi

    Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4

    Science

    (2011)
  • M.F. Krummel et al.

    CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation

    J. Exp. Med.

    (1995)
  • H.S. Kuehn

    Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4

    Science

    (2014)
  • K. Wing

    CTLA-4 control over Foxp3+ regulatory T cell function

    Science

    (2008)
  • M.E. Keir et al.

    PD-1 and its ligands in tolerance and immunity

    Annu. Rev. Immunol.

    (2008)
  • M.E. Keir

    Tissue expression of PD-L1 mediates peripheral T cell tolerance

    J. Exp. Med.

    (2006)
  • Y. Latchman

    PD-L2 is a second ligand for PD-1 and inhibits T cell activation

    Nat. Immunol.

    (2001)
  • G.J. Freeman

    Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation

    J. Exp. Med.

    (2000)
  • A.M. Luoma

    Molecular pathways of Colon inflammation induced by Cancer immunotherapy

    Cell

    (2020)
  • J.A. Bluestone et al.

    Tolerance in the age of immunotherapy

    N. Engl. J. Med.

    (2020)
  • J.R. Ingram

    Anti-CTLA-4 therapy requires an Fc domain for efficacy

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

    (2018)
  • T.R. Simpson

    Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma

    J. Exp. Med.

    (2013)
  • M.J. Selby

    Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells

    Cancer Immunol. Res.

    (2013)
  • Cited by (13)

    • The changing clinical spectrum of endocrine adverse events in cancer immunotherapy

      2022, Trends in Endocrinology and Metabolism
      Citation Excerpt :

      Although the mechanisms of ICI toxicity have not been completely clarified, inhibition of immune checkpoints may reduce immune tolerance to autoantigens, resulting in the development of autoimmunity disorders [22]. Indeed, the toxicity of ICIs confirms the crucial role of the CTLA-4 and PD-1/PD-L1 pathways in human immune homeostasis and tolerance [3]. Diverse pathophysiological mechanisms have been suggested to underlie the onset of IRAEs, and this could explain their great heterogeneity [23] both in terms of severity and the organs involved (e.g., skin, lung, liver, bowel, and endocrine glands) [24].

    View all citing articles on Scopus
    View full text