Diabetes Metab J.  2023 Nov;47(6):757-766. 10.4093/dmj.2023.0072.

Immune-Checkpoint Inhibitors-Induced Type 1 Diabetes Mellitus: From Its Molecular Mechanisms to Clinical Practice

Affiliations
  • 1Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
  • 2Asan Diabetes Center, Asan Medical Center, Seoul, Korea

Abstract

With the increasing use of immune-checkpoint inhibitors (ICIs), such as anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and anti-programmed cell death-1 (PD-1), for the treatment of malignancies, cases of ICI-induced type 1 diabetes mellitus (ICI-T1DM) have been reported globally. This review focuses on the features and pathogenesis of this disease. T1DM is an immune-related adverse event that occurs following the administration of anti-PD-1 or anti-programmed death ligand-1 (PDL1) alone or in combination with anti-CTLA-4. More than half of the reported cases presented as abrupt-onset diabetic ketoacidosis. The primary mechanism of ICI-T1DM is T-cell stimulation, which results from the loss of interaction between PD-1 and PD-L1 in pancreatic islet. The similarities and differences between ICI-T1DM and classical T1DM may provide insights into this disease entity. ICI-T1DM is a rare but often life-threatening medical emergency that healthcare professionals and patients need to be aware of. Early detection of and screening for this disease is imperative. At present, the only known treatment for ICI-T1DM is insulin injection. Further research into the mechanisms and risk factors associated with ICI-T1DM development may contribute to a better understanding of this disease entity and the identification of possible preventive strategies.

Keyword

Diabetes mellitus, type 1; Immune checkpoint inhibitors; Neoplasms

Figure

  • Fig. 1. Effect of immune-checkpoint inhibitors on T lymphocytes. Anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA4) blocking and anti-programmed cell death-1 (PD-1) or antiprogrammed death ligand-1 (PD-L1) blocking restore pro-activatory signaling and result in effective antitumor T lymphocyte responses. TCR, T-cell receptor; MHC, major histocompatibility complex.

  • Fig. 2. Pathogenesis of classic type 1 diabetes mellitus (T1DM) and immune-checkpoint inhibitor-induced T1DM (ICI-T1DM). In T1DM, inappropriate immune reaction can lead to an autoimmune response by autoreactive T-cells. The programmed cell death-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitory pathway plays a fundamental role in the maintenance of immune tolerance, thus, dysregulation of the PD-1/PD-L1 is an important pathogenesis of classical T1DM. Immunotherapeutic inhibition of PD-1 or its receptor PD-L1 to target cancer cells, involves autoimmune reactions which resembles the pathogenesis of T1DM. With immune-checkpoint inhibitors, unintended immune and autoimmune responses including those against pancreatic islets might be activated. HLA, human leukocyte antigen.


Reference

1. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. 2020; 11:3801.
Article
2. Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019; 2:e192535.
Article
3. Vilgelm AE, Johnson DB, Richmond A. Combinatorial approach to cancer immunotherapy: strength in numbers. J Leukoc Biol. 2016; 100:275–90.
Article
4. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA Oncol. 2019; 5:1411–20.
Article
5. Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol. 2022; 19:254–67.
Article
6. Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, et al. Defining ‘T cell exhaustion’. Nat Rev Immunol. 2019; 19:665–74.
Article
7. Philip M, Fairchild L, Sun L, Horste EL, Camara S, Shakiba M, et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature. 2017; 545:452–6.
Article
8. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018; 8:1069–86.
Article
9. Barton BM, Xu R, Wherry EJ, Porrett PM. Pregnancy promotes tolerance to future offspring by programming selective dysfunction in long-lived maternal T cells. J Leukoc Biol. 2017; 101:975–87.
Article
10. Shim YJ, Khedraki R, Dhar J, Fan R, Dvorina N, Valujskikh A, et al. Early T cell infiltration is modulated by programed cell death-1 protein and its ligand (PD-1/PD-L1) interactions in murine kidney transplants. Kidney Int. 2020; 98:897–905.
Article
11. Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 2021; 14:45.
Article
12. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 2018; 8:86.
Article
13. Das S, Johnson DB. Immune-related adverse events and antitumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019; 7:306.
Article
14. Quach HT, Dewan AK, Davis EJ, Ancell KK, Fan R, Ye F, et al. Association of anti-programmed cell death 1 cutaneous toxic effects with outcomes in patients with advanced melanoma. JAMA Oncol. 2019; 5:906–8.
Article
15. Eggermont AM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, et al. Association between immune-related adverse events and recurrence-free survival among patients with stage III melanoma randomized to receive pembrolizumab or placebo: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2020; 6:519–27.
Article
16. Yoest JM. Clinical features, predictive correlates, and pathophysiology of immune-related adverse events in immune checkpoint inhibitor treatments in cancer: a short review. Immunotargets Ther. 2017; 6:73–82.
Article
17. Passat T, Touchefeu Y, Gervois N, Jarry A, Bossard C, Bennouna J. Physiopathological mechanisms of immune-related adverse events induced by anti-CTLA-4, anti-PD-1 and anti-PD-L1 antibodies in cancer treatment. Bull Cancer. 2018; 105:1033–41.
18. Johnson DB, McDonnell WJ, Gonzalez-Ericsson PI, Al-Rohil RN, Mobley BC, Salem JE, et al. A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat Med. 2019; 25:1243–50.
Article
19. Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016; 7:10391.
Article
20. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med. 2014; 6:230. ra45.
Article
21. Wei SC, Meijers WC, Axelrod ML, Anang NA, Screever EM, Wescott EC, et al. A genetic mouse model recapitulates immune checkpoint inhibitor-associated myocarditis and supports a mechanism-based therapeutic intervention. Cancer Discov. 2021; 11:614–25.
Article
22. Luoma AM, Suo S, Williams HL, Sharova T, Sullivan K, Manos M, et al. Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell. 2020; 182:655–71.
Article
23. Andrews MC, Duong CP, Gopalakrishnan V, Iebba V, Chen WS, Derosa L, et al. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med. 2021; 27:1432–41.
Article
24. Roep BO, Thomaidou S, van Tienhoven R, Zaldumbide A. Type 1 diabetes mellitus as a disease of the β-cell (do not blame the immune system?). Nat Rev Endocrinol. 2021; 17:150–61.
Article
25. Nguyen C, Varney MD, Harrison LC, Morahan G. Definition of high-risk type 1 diabetes HLA-DR and HLA-DQ types using only three single nucleotide polymorphisms. Diabetes. 2013; 62:2135–40.
Article
26. Herold KC, Bundy BN, Long SA, Bluestone JA, DiMeglio LA, Dufort MJ, et al. An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. N Engl J Med. 2019; 381:603–13.
Article
27. Carvalho T. FDA approves first drug to delay type 1 diabetes. Nat Med. 2023; 29:280.
Article
28. Nerup J, Platz P, Andersen OO, Christy M, Lyngsoe J, Poulsen JE, et al. HL-A antigens and diabetes mellitus. Lancet. 1974; 2:864–6.
Article
29. Barrett JC, Clayton DG, Concannon P, Akolkar B, Cooper JD, Erlich HA, et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet. 2009; 41:703–7.
Article
30. van Lummel M, van Veelen PA, de Ru AH, Janssen GM, Pool J, Laban S, et al. Dendritic cells guide islet autoimmunity through a restricted and uniquely processed peptidome presented by high-risk HLA-DR. J Immunol. 2016; 196:3253–63.
Article
31. van Lummel M, van Veelen PA, de Ru AH, Pool J, Nikolic T, Laban S, et al. Discovery of a selective islet peptidome presented by the highest-risk HLA-DQ8trans molecule. Diabetes. 2016; 65:732–41.
32. Kim KY, Kim MS, Lee YJ, Lee YA, Lee SY, Shin CH, et al. Glutamic acid decarboxylase and tyrosine phosphatase-related islet antigen-2 positivity among children and adolescents with diabetes in Korea. Diabetes Metab J. 2022; 46:948–52.
Article
33. Vella A, Cooper JD, Lowe CE, Walker N, Nutland S, Widmer B, et al. Localization of a type 1 diabetes locus in the IL2RA/CD25 region by use of tag single-nucleotide polymorphisms. Am J Hum Genet. 2005; 76:773–9.
Article
34. Mourad D, Azar NS, Eid AA, Azar ST. Immune checkpoint inhibitor-induced diabetes mellitus: potential role of T cells in the underlying mechanism. Int J Mol Sci. 2021; 22:2093.
Article
35. Won TJ, Jung YJ, Kwon SJ, Lee YJ, Lee DI, Min H, et al. Forced expression of programmed death-1 gene on T cell decreased the incidence of type 1 diabetes. Arch Pharm Res. 2010; 33:1825–33.
Article
36. Roep BO, Wheeler DC, Peakman M. Antigen-based immune modulation therapy for type 1 diabetes: the era of precision medicine. Lancet Diabetes Endocrinol. 2019; 7:65–74.
Article
37. Okazaki T, Honjo T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006; 27:195–201.
Article
38. Wang CJ, Chou FC, Chu CH, Wu JC, Lin SH, Chang DM, et al. Protective role of programmed death 1 ligand 1 (PD-L1) in nonobese diabetic mice: the paradox in transgenic models. Diabetes. 2008; 57:1861–9.
39. Osum KC, Burrack AL, Martinov T, Sahli NL, Mitchell JS, Tucker CG, et al. Interferon-gamma drives programmed death-ligand 1 expression on islet β cells to limit T cell function during autoimmune diabetes. Sci Rep. 2018; 8:8295.
Article
40. Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H, et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med. 2003; 198:63–9.
Article
41. Pauken KE, Jenkins MK, Azuma M, Fife BT. PD-1, but not PD-L1, expressed by islet-reactive CD4+ T cells suppresses infiltration of the pancreas during type 1 diabetes. Diabetes. 2013; 62:2859–69.
Article
42. Tsutsumi Y, Jie X, Ihara K, Nomura A, Kanemitsu S, Takada H, et al. Phenotypic and genetic analyses of T-cell-mediated immunoregulation in patients with type 1 diabetes. Diabet Med. 2006; 23:1145–50.
43. Al-Hakami A. Serum sCTLA-4 level is not associated with type 1 diabetes or the coexistence of autoantibodies in children and adolescent patients from the southern region of Saudi Arabia. Auto Immun Highlights. 2020; 11:19.
Article
44. Colli ML, Hill JL, Marroqui L, Chaffey J, Dos Santos RS, Leete P, et al. PD-L1 is expressed in the islets of people with type 1 diabetes and is up-regulated by interferons-α and-γ via IRF1 induction. EBioMedicine. 2018; 36:367–75.
Article
45. Li X, Zhong T, Tang R, Wu C, Xie Y, Liu F, et al. PD-1 and PD-L1 expression in peripheral CD4/CD8+ T cells is restored in the partial remission phase in type 1 diabetes. J Clin Endocrinol Metab. 2020; 105:dgaa130.
Article
46. Kapke J, Shaheen Z, Kilari D, Knudson P, Wong S. Immune checkpoint inhibitor-associated type 1 diabetes mellitus: case series, review of the literature, and optimal management. Case Rep Oncol. 2017; 10:897–909.
Article
47. Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA, et al. Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors. Diabetes. 2018; 67:1471–80.
Article
48. de Filette JM, Pen JJ, Decoster L, Vissers T, Bravenboer B, Van der Auwera BJ, et al. Immune checkpoint inhibitors and type 1 diabetes mellitus: a case report and systematic review. Eur J Endocrinol. 2019; 181:363–74.
Article
49. Hu H, Zakharov PN, Peterson OJ, Unanue ER. Cytocidal macrophages in symbiosis with CD4 and CD8 T cells cause acute diabetes following checkpoint blockade of PD-1 in NOD mice. Proc Natl Acad Sci U S A. 2020; 117:31319–30.
Article
50. Yoneda S, Imagawa A, Hosokawa Y, Baden MY, Kimura T, Uno S, et al. T-lymphocyte infiltration to islets in the pancreas of a patient who developed type 1 diabetes after administration of immune checkpoint inhibitors. Diabetes Care. 2019; 42:e116–8.
Article
51. Perdigoto AL, Deng S, Du KC, Kuchroo M, Burkhardt DB, Tong A, et al. Immune cells and their inflammatory mediators modify β cells and cause checkpoint inhibitor-induced diabetes. JCI Insight. 2022; 7:e156330.
Article
52. Quandt Z, Young A, Anderson M. Immune checkpoint inhibitor diabetes mellitus: a novel form of autoimmune diabetes. Clin Exp Immunol. 2020; 200:131–40.
Article
53. Rui J, Deng S, Arazi A, Perdigoto AL, Liu Z, Herold KC. β Cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice. Cell Metab. 2017; 25:727–38.
Article
54. Tachibana M, Imagawa A. Type 1 diabetes related to immune checkpoint inhibitors. Best Pract Res Clin Endocrinol Metab. 2022; 36:101657.
Article
55. Baden MY, Imagawa A, Abiru N, Awata T, Ikegami H, Uchigata Y, et al. Characteristics and clinical course of type 1 diabetes mellitus related to anti-programmed cell death-1 therapy. Diabetol Int. 2018; 10:58–66.
Article
56. Lo Preiato V, Salvagni S, Ricci C, Ardizzoni A, Pagotto U, Pelusi C. Diabetes mellitus induced by immune checkpoint inhibitors: type 1 diabetes variant or new clinical entity?: review of the literature. Rev Endocr Metab Disord. 2021; 22:337–49.
Article
57. Kotwal A, Haddox C, Block M, Kudva YC. Immune checkpoint inhibitors: an emerging cause of insulin-dependent diabetes. BMJ Open Diabetes Res Care. 2019; 7:e000591.
Article
58. Chen X, Affinati AH, Lee Y, Turcu AF, Henry NL, Schiopu E, et al. Immune checkpoint inhibitors and risk of type 1 diabetes. Diabetes Care. 2022; 45:1170–6.
Article
59. Akturk HK, Kahramangil D, Sarwal A, Hoffecker L, Murad MH, Michels AW. Immune checkpoint inhibitor-induced type 1 diabetes: a systematic review and meta-analysis. Diabet Med. 2019; 36:1075–81.
Article
60. Zezza M, Kosinski C, Mekoguem C, Marino L, Chtioui H, Pitteloud N, et al. Combined immune checkpoint inhibitor therapy with nivolumab and ipilimumab causing acute-onset type 1 diabetes mellitus following a single administration: two case reports. BMC Endocr Disord. 2019; 19:144.
Article
61. Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, et al. Treatment-related adverse events of PD-1 and PD-L1 inhibitors in clinical trials: a systematic review and meta-analysis. JAMA Oncol. 2019; 5:1008–19.
Article
62. Marmura H, Tremblay PF, Getgood AM, Bryant DM. The knee injury and osteoarthritis outcome score does not have adequate structural validity for use with young, active patients with ACL tears. Clin Orthop Relat Res. 2022; 480:1342–50.
Article
63. Takada S, Hirokazu H, Yamagishi K, Hideki S, Masayuki E. Predictors of the onset of type 1 diabetes obtained from real-world data analysis in cancer patients treated with immune checkpoint inhibitors. Asian Pac J Cancer Prev. 2020; 21:1697–9.
Article
64. Wright JJ, Salem JE, Johnson DB, Lebrun-Vignes B, Stamatouli A, Thomas JW, et al. Increased reporting of immune checkpoint inhibitor-associated diabetes. Diabetes Care. 2018; 41:e150–1.
Article
65. Liu J, Shi Y, Liu X, Zhang D, Zhang H, Chen M, et al. Clinical characteristics and outcomes of immune checkpoint inhibitor-induced diabetes mellitus. Transl Oncol. 2022; 24:101473.
Article
66. Qiu J, Luo S, Yin W, Guo K, Xiang Y, Li X, et al. Characterization of immune checkpoint inhibitor-associated fulminant type 1 diabetes associated with autoantibody status and ethnic origin. Front Immunol. 2022; 13:968798.
Article
67. Dougan M, Pietropaolo M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J Clin Invest. 2020; 130:51–61.
Article
68. Imagawa A, Hanafusa T, Miyagawa J, Matsuzawa Y. A novel subtype of type 1 diabetes mellitus characterized by a rapid onset and an absence of diabetes-related antibodies. Osaka IDDM Study Group. N Engl J Med. 2000; 342:301–7.
Article
69. Shiba M, Inaba H, Ariyasu H, Kawai S, Inagaki Y, Matsuno S, et al. Fulminant type 1 diabetes mellitus accompanied by positive conversion of anti-insulin antibody after the administration of anti-CTLA-4 antibody following the discontinuation of anti-PD-1 antibody. Intern Med. 2018; 57:2029–34.
Article
70. Lee M, Jeong K, Park YR, Rhee Y. Increased risk of incident diabetes after therapy with immune checkpoint inhibitor compared with conventional chemotherapy: a longitudinal trajectory analysis using a tertiary care hospital database. Metabolism. 2023; 138:155311.
Article
71. Hong AR, Yoon JH, Kim HK, Kang HC. Immune checkpoint inhibitor-induced diabetic ketoacidosis: a report of four cases and literature review. Front Endocrinol (Lausanne). 2020; 11:14.
Article
72. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. 2. Classification and diagnosis of diabetes: standards of care in diabetes-2023. Diabetes Care. 2023; 46(Suppl 1):S19–40.
73. Smith CJ, Almodallal Y, Jatoi A. Rare adverse events with programmed death-1 and programmed death-1 ligand inhibitors: justification and rationale for a systematic review. Curr Oncol Rep. 2021; 23:86.
74. Zhao Z, Wang X, Bao XQ, Ning J, Shang M, Zhang D. Autoimmune polyendocrine syndrome induced by immune checkpoint inhibitors: a systematic review. Cancer Immunol Immunother. 2021; 70:1527–40.
Article
75. Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, et al. American Association of Clinical Endocrinology clinical practice guideline: developing a diabetes mellitus comprehensive care plan-2022 update. Endocr Pract. 2022; 28:923–1049.
Article
76. Holt RI, DeVries JH, Hess-Fischl A, Hirsch IB, Kirkman MS, Klupa T, et al. The management of type 1 diabetes in adults: a consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. 2021; 64:2609–52.
Article
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr