Review
Immune checkpoint inhibitors and diabetes: Mechanisms and predictors

https://doi.org/10.1016/j.diabet.2020.09.003Get rights and content

Abstract

The emergence of immune checkpoint inhibitors in the arsenal of cancer immunotherapy was a breakthrough which provided hope to many cancer patients. However, not long has passed since their discovery that some adverse effects were associated with these promising therapeutic agents. Immune checkpoint inhibitors dysregulate host immunity and may precipitate autoimmune diseases including diabetes mellitus. In this review, we go beyond the case reports towards understanding the underlying mechanisms by which Programmed cell death 1 (PD-1) and Programmed death ligand-1 (PD-L1) inhibitors precipitate diabetes. We discuss the role of PD-1/PD-L1 in autoimmunity and the use of mice models to describe their involvement in diabetes. We also reviewed the genetic anomalies in PD-1/PD-L1genes and their link to diabetes. Finally, we present the studies conducted to identify patients at risk of developing autoimmune diseases as an adverse effect for PD-1/PD-L1 use. Understanding these issues can guide researchers to find a way to circumvent the autoimmune adverse reactions seen with PD-1/PD-L1 inhibitors without affecting their antitumor activity.

Section snippets

PD-1/PD-L1 inhibitors: the breakthrough and the glitch

Immune checkpoint inhibitors (CPI) include inhibitors for PD-1 and its ligand (PD-L1) [14], [15], [16], [17], [18]. PD-1 is normally expressed on the surface of activated T-cells enabling them to interact with PD-L1 and other receptors on the surface of antigen-presenting cells as well as tumour cells [19]. This interaction will result in the neutralization of T-cells and unresponsiveness. The neutralization of T-cells will render them unresponsive towards tumour cells allowing them to evade

Role of immunity in diabetes: the sabotaging traitor

Immunity plays a key role in the pathogenesis of diabetes. Both T1D and T2D are supported by improper host immune response. T1D is precipitated due to improper insulin secretion by pancreatic β-cells because of its T cell-mediated destruction [22]. The second arm of the immune system i.e. B cells are also involved in this process through the secretion of autoantibodies. There are 4 main autoantibodies involved in the destruction of β-cells and the development of T1D namely Islet Cell Antibodies

T1D as an adverse effect for checkpoint inhibitors: the hint

Although clinical trials of PD-1 and PD-L1 inhibitors showed very promising therapeutic potentials as anti-tumours, they revealed immune-related adverse effects disturbing the metabolic homeostasis [27], [28]. One of the adverse effects that has been reported from animal and clinical studies is T1D [29], [30], [31]. Although diabetes development secondary to CPI is not very common (0.9% in the 6 year period examined by Stamatouli et al. 2018), however, this number is still considered clinically

PD-1/PD-L1 in type 1 diabetes: A researcher and mouse game

Given the importance of highlighting the cellular and molecular mechanisms behind the onset of secondary type 1 diabetes observed with immune checkpoint inhibition, several experimental animal models were developed. The mechanism behind T1D development as a secondary result to immune checkpoint inhibition, is thought to be the same as the antitumor mechanism of these therapeutic agents [27]. A finding that makes it extremely difficult to avoid diabetic ketoacidosis onset with the use of

The genetics story

The susceptibility to secondary T1D can be deduced by genotyping, with the largest influence imparted by the HLA genotype. It has been shown that patients heterozygous for the MHC I HLA loci show better outcomes when treated with immune checkpoint inhibitors [52]. Similarly, spontaneous T1D is associated with HLA-DR4, DR3, DQ2 and DQ8 genotypes. From these, HLA-DR4 genotype was shown to be the predominant genotype in T1D as a CPI adverse effect as it was present in 42% of the patients (χ2 test,

Biomarkers for secondary insulin dependent diabetes: the tattletales

In an effort to discover biomarkers for the high risk group for predisposition to T1D when treated with CPI, a recent clinical study has reported an association between T1D and elevated levels of circulating peripheral T helper cells (Tph) that are negative for Csingle bondXsingle bondC motif chemokine receptor type 5 and positive for programmed cell death protein 1-positive (CXCR5PD-1hi). This was found both before and following the diagnosis of T1D [63]. The Tph CXCR5PD-1hi cells can be used as a biomarker for

CPIs and type 2 diabetes: two for the price of one

It is intuitive that CPI are associated with several autoimmune disorders including T1D because they are involved in immune regulation. However, the incidence of T1D tempted researchers to investigate if CPI can predispose T2D as well. In a retrospective study, Gauci et al. reported occurrence of type 1 and type 2 diabetes in patients treated with PD-1 and CTLA-4 inhibitors [27], [66]. Their study included all the patients that attended the Oncodermatology clinics of Saint Louis Hospital in

Conclusion

Immune checkpoint inhibitors are being used in combinations to give a better antitumor effects, however, a combination therapy might be associated with a higher risk of autoimmune diabetes development [32]. While the incidence of diabetes is a rare side effects for CPI monotherapy, CPIs are usually used in combinations which increases the incidence of this adverse effect. This fact underscores the importance of the identification of biomarkers that can predict the susceptibility of certain

Author contributions

NY and MN performed the literature review and wrote the manuscript. GD helped in the revision of the review. AAE conceived and designed the review article. All authors approved the final version of the review.

Role of the funding source

This content is solely the responsibility of the authors. The funding sources had no involvement in any aspect of the research.

Conflict of interest

None.

Acknowledgments

NY and MN are supported by a doctoral fellowship from the American University of Beirut, Faculty of Medicine. AAE is supported by the American University of Beirut Medical Practice Plan regular research grant.

References (71)

  • E. Nolan et al.

    Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer

    Sci Transl Med

    (2017)
  • S. Marwitz et al.

    Epigenetic modifications of the immune-checkpoint genes CTLA4 and PDCD1 in non-small cell lung cancer results in increased expression

    Clin Epigenetics

    (2017)
  • P. Zheng et al.

    Human cancer immunotherapy with PD-1/PD-L1 blockade

    Biomark Cancer

    (2015)
  • K. Cassady et al.

    Regulation of GVHD and GVL activity via PD-L1 interaction with PD-1 and CD80

    Front Immunol

    (2018)
  • B.T. Fife et al.

    Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways

    Immunol Rev

    (2008)
  • R. Fujisawa et al.

    Low programmed cell death-1 (PD-1) expression in peripheral CD4+ T cells in Japanese patients with autoimmune type 1 diabetes

    Clin Exp Immunol

    (2015)
  • B.T. Fife et al.

    Interactions between programmed death-1 and programmed death ligand-1 promote tolerance by blocking the T cell receptor-induced stop signal

    Nat Immunol

    (2009)
  • W. Zou et al.

    Inhibitory B7-family molecules in the tumour microenvironment

    Nat Rev Immunol

    (2008)
  • E.N. Rozali et al.

    Programmed death ligand 2 in cancer-induced immune suppression

    Clin Dev Immunol

    (2012)
  • A. Ribas et al.

    Cancer immunotherapy using checkpoint blockade

    Science

    (2018)
  • A. Ribas

    Tumor immunotherapy directed at PD-1

    N Engl J Med

    (2012)
  • T. Okazaki et al.

    PD-1 and PD-1 ligands: from discovery to clinical application

    Int Immunol

    (2007)
  • L. Chen et al.

    Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future

    J Clin Invest

    (2015)
  • H.M. Zarour

    Reversing T-cell dysfunction and exhaustion in cancer

    Clin Cancer Res

    (2016)
  • S.M. Ferrari et al.

    Thyroid disorders induced by checkpoint inhibitors

    Rev Endocr Metab Disord

    (2018)
  • K.C. Herold et al.

    Type 1 diabetes: translating mechanistic observations into effective clinical outcomes

    Nat Rev Immunol

    (2013)
  • A.G. Ziegler et al.

    Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children

    JAMA

    (2013)
  • M.D. Pescovitz et al.

    Rituximab, B-Lymphocyte depletion, and preservation of beta-cell function

    N Engl J Med

    (2009)
  • J.C. Pickup

    Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes

    Diabetes Care

    (2004)
  • A.M. Stamatouli et al.

    Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors

    Diabetes

    (2018)
  • M.A. Postow et al.

    Immune-related adverse events associated with immune checkpoint blockade

    N Engl J Med

    (2018)
  • M.J.I. Ansari et al.

    The programmed Death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice

    J Exp Med

    (2003)
  • J. Hughes et al.

    Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy

    Diabetes Care

    (2015)
  • J. Martin-Liberal et al.

    Anti-programmed cell death-1 therapy and insulin-dependent diabetes: a case report

    Cancer Immunol Immunother

    (2015)
  • H.K. Akturk et al.

    Immune checkpoint inhibitor‐induced Type 1 diabetes: a systematic review and meta‐analysis

    Diabet Med

    (2019)
  • Cited by (10)

    View all citing articles on Scopus
    1

    The Authors Equally Contributed to the Write-Up of this Review Manuscript.

    View full text