Skip to main content

CASE REPORT article

Front. Public Health, 16 June 2022
Sec. Clinical Diabetes

Anti-programmed Cell Death Protein-1 Therapy in Intrahepatic Cholangiocarcinoma Induced Type 1 Diabetes: A Case Report and Literature Review

\nZhi-Kai Zheng,Zhi-Kai Zheng1,2Jiong-Liang Wang,Jiong-Liang Wang1,2Wen-Xuan Li,Wen-Xuan Li1,2Tian-Qing Wu,Tian-Qing Wu1,2Min-Shan Chen,Min-Shan Chen1,2Zhong-Guo Zhou,
Zhong-Guo Zhou1,2*
  • 1Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
  • 2Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China

Immune checkpoint inhibitors, widely used in the treatment of malignancies, can improve the prognosis of patients, while it also can induce various immune-related adverse events, and type 1 diabetes induced by anti-programmed cell death protein-1 is a rare but severe complication. Here we reported a case of type 1 diabetes induced by anti-PD-1 which was to treat intrahepatic cholangiocarcinoma. The case was a 61-year-old female who developed diabetes and ketoacidosis symptoms at the 16th week after anti-PD-1 therapy. Her blood glucose was 30.32 mmol/L, HBA1c was 8.10%, and C-peptide was <0.10 ng/ml. The patient was diagnosed as fulminant type 1 diabetes mellitus complicated with ketoacidosis induced by anti-PD-1, and was treated with massive fluid rehydration, intravenous infusion of insulin and correction of acid-base electrolyte disorder. Hepatectomy was performed after stabilization, and the patient was treated with long-term insulin. Through the case report and literature review, this study aims to improve oncologists' understanding of anti-PD-1 induced type 1 diabetes, so as to make early diagnosis and treatment of the complications and ensure medical safety.

Introduction

Immune checkpoint inhibitors (ICIs), which have been widely applied to patients with advanced malignancies, can improve the prognosis of patients (13). It can enhance the anti-tumor immune response by blocking immune checkpoints (ICs) (4). As the representative of ICIs, anti-programmed cell death protein 1 (anti-PD-1) has been studied in animals and in several clinical trials. It was approved by the Food and Drug Administration (FDA) for the treatment of types of cancers, such as squamous cell carcinoma of head and neck, non-small cell lung cancer, renal carcinoma, urothelial carcinoma, melanoma, Hodgkin's lymphoma, liver cancer, colorectal cancer and so on (5, 6). In addition, immunotherapy can also be used for other advance solid tumors with limited therapies, such as intrahepatic cholangiocarcinoma, and it has achieved great success (7, 8).

However, ICIs can induce various immune related adverse events (irAEs) including pneumonitis, hepatitis, colitis, dermatitis and endocrinopathies (9, 10). Endocrinopathies are common complications, including diabetes mellitus, hypophysitis, thyroid dysfunction, primary adrenal insufficiency (11). Although the incidence of immune checkpoint inhibitor-induced diabetes is low, it may lead to life-threatening diabetic ketoacidosis. So it's necessary for us to understand the clinical manifestation, examination and therapies of the immune checkpoint inhibitor-induced diabetes. In this report, we present a patient with intrahepatic cholangiocarcinoma who had anti-PD-1-induced type 1 diabetes, in order to improve oncologists' understanding of type 1 diabetes induced by anti-PD-1.

Case Presentation

The patient, a 61-year-old female with no diabetes mellitus history and family history of diabetes, whose HBsAg, HBeAb, and HBcAb were all positive while the anti-HCV was negative, was diagnosis as intrahepatic cholangiocarcinoma by pathology in March 25th, 2021. However, the hepatic mass was unresectable. Hepatic arterial infusion chemotherapy (FOLFOX protocol: Oxaliplatin 130 mg/m2, Calcium levofolinate 200 mg/m2, 5-fluorouracil 400 mg/m2) combined with anti-PD-1 immunotherapy (Tislelizumab 200 mg) was performed from March 25, 2021 to June 17, 2021 for 3 cycles that every 3 weeks was a cycle. No abnormal blood glucose and HBA1c were detected during medication. After that the Magnetic resonance imagination and CT examination on July 8, 2021 showed that the hepatic mass was smaller than before, which meant that the patient was admitted to the hospital for liver cancer resection. However, on the second day after admission, the patient felt thirsty, nauseous and somnolent. The laboratory test (Table 1) was as following: glucose: 30.32 mmol/L, HBA1c: 7.6%, pH: 7.18, PaCO2: 26.6 mmHg, HCO3-: 9.7 mmol/L, lactic acid: 2.9 mmol/L, urinary glucose: 4+, urinary ketone: 4+, GAD: -, ICA: -, IAA: -, IA-2A: -. Considering her clinical manifestation and examination, type 1 diabetes accompanying with ketoacidosis induced by anti-PD-1 was diagnosed. Intravenous massive fluid infusion, infusion of insulin and correction of acid-base electrolyte disorder were given. After that, the diabetes related laboratory test (Table 2) was reexamined and as following: Blood glucose: 20.14 mmol/L, HBA1c: 7.60%, insulin (0 h): 9.03 mU/L, (1 h) 55.34 mU/L, (2 h) 37.52 mU/L, C-peptide (0 h): 0.09 ng/ mL, (1 h): 0.08 ng/ mL, (2 h): 0.08 ng/ml. After consultation with the endocrinology department, the insulin dose was adjusted for several times. Finally, insulin aspartate (4iu in the morning, 6iu at noon, 8iu in the evening I.H. TID) and insulin glargine (10iu at 10 PM I.H. QN) were administered, and the control of blood glucose was stable. After 2 weeks, the patient was admitted to the hospital again for intrahepatic cholangiocarcinoma resection, and the operation was successful. One week after surgery, the patient was discharged safely and insulin was continued to treat diabetes. One month after the operation, the patient returned to the hospital for further consultation. There was no abnormality in liver tumor-associated antigen and blood glucose and no tumor recurrence showed by magnetic resonance imagination. However, the patient was dependent on insulin therapy.

TABLE 1
www.frontiersin.org

Table 1. The laboratory test.

TABLE 2
www.frontiersin.org

Table 2. The reexamined laboratory test.

Discussion

The Incidence of Anti-PD-1 Diabetes

Anti-PD-1 induced diabetes is a relatively rare complication, whose incidence ranges from 0.2 to 2% (1, 2, 1215). The meta-analysis by Akturk et al. (16) and the study by Youssef et al. (15) both indicated a current incidence of 0.9%. In addition, different kinds of anti-PD-1 lead to different rates of diabetes. Several studies showed that the incidence of diabetes induced by Nivolumab is 0.9%, higher than that of Pebolizumab (0.2%) (9, 17, 18). Due to the loss of blood glucose monitoring and the inobvious early symptoms of the disease, the reported incidence is lower than the actual. And as the anti-PD-1 is initiated at an earlier disease stage and to a greater extent, the incidence of immunotherapy-related diabetes will increase gradually (1, 4).

The Pathogenesis of Anti-PD-1 Diabetes

PD-1 receptor is a type 1 transmembrane glycoprotein with a molecular weight of 50–55 kDa, belonging to the immunoglobulin superfamily, whose ligands are PD-L1 and PD-L2 (19, 20). The mechanism of anti-PD-1 leading to diabetes is still unclear, and it may be associated with activated T cells. PD-1 exists on the surface of T cells, and it can suppress the activation and proliferation of T cells through inhibiting the PI3K/Akt pathway to block the uptake of glucose and gluconeogenesis by binding with ligands on the surface of immune cells, islet cells and other cells, which results in immune tolerance (11, 1823). By blocking the PD-1/PD-L1 and PD-L2 pathways, anti-PD-1 blocks the inhibitory signal of T cells, leading to the proliferation and activation of islet cell-specific T cells, which leads to the destruction of islet cells and the occurrence of diabetes (21, 24). This has been demonstrated in several mouse models (2, 15, 20, 21, 25). The mechanism of anti-PD-1 diabetes still needs further study.

The Clinical Manifestations of Anti-PD-1 Diabetes

Similar to traditional type 1 diabetes, diabetes induced by anti-PD-1 may present symptoms and signs of hyperglycemia, such as polyuria, polydipsia, polyphagia (20). Some patients may be accompanied with diabetic ketoacidosis (DKA), which presents as nausea, vomiting, abdominal pain, dyspnea, somnolence and even coma (3). The incidence of DKA is very high, ranging from 62.1 to 81% (11, 13, 16, 21, 23). DKA is always the reason for diagnosis, which greatly increases the risk of death, which is also the reason why physicians need to be vigilant against diabetes induced by anti-PD-1 (11). In addition, patients may have no symptoms and be diagnosed with hyperglycemia only by occasional blood glucose monitoring. In this case, the patient's symptoms were obvious and were successfully controlled after active treatment, which indicates the importance of early identification and therapy of diabetes.

The Laboratory and Imaging Examination of Anti-PD-1 Diabetes

In order to timely identify anti-PD-1 diabetes and prevent the occurrence of ketoacidosis, it is necessary to find some relative biomarkers or indicators.

Blood Glucose

The blood glucose levels of patients with anti-PD-1 diabetes were elevated, which were 33.4 ± 11.5 (13.7–67.3) mmol/L [602 ± 207 (246–1,211) mg/dl] (16). The time to onset varied from 1 to 52 weeks (10, 11, 13, 14, 16, 18, 26). In this case, the patient's blood glucose level was very high at onset (30.32 mmol/L), and the onset time was 16 weeks after treatment with anti-PD-1, which was consistent with literature reports. Due to the uncertainty of blood glucose level, onset time and high incidence of DKA, blood glucose monitoring must be part of routine monitoring before and during anti-PD-1 treatment (4, 20). As for the frequency of monitoring, Akturk et al. (16) pointed out that 71% of patients developed diabetes within 3 months after anti-PD-1 treatment, so the frequency of blood glucose detection should be appropriately increased at the beginning of treatment and gradually decreased after 3–4 months (18).

Hemoglobin A1c

Hemoglobin A1c (HbA1c), a measure of average blood glucose over the preceding 3 months, is a common indicator for diabetes monitoring. Different from traditional diabetes, HbA1c levels in patients with anti-PD-1 diabetes were mostly close to normal or slightly elevated, ranging from 5.8 to 11.4% (1, 4, 5, 18, 21, 22, 27). In this case, the patient's HbA1c was 7.6%. Since the acute onset of diabetes induced by anti-PD-1, the early HbA1c level is mostly normal, which might not be a good screening indicator (11). Nevertheless, continuous monitoring of HbA1c is significant for patients to minimize the risk of diabetes development and adjust the treatment regimen (15, 22). Routine monitoring of HbA1c after anti-PD-1 treatment is still of importance.

C-Peptide

C-peptide, which is secreted by islet β cells, is an indicator of islet β cell function and is not influenced by exogenous insulin. In patients with anti-PD-1 diabetes, C-peptide levels might be low or even absent, which suggests the rapid destruction of β cells (1, 3, 4, 14, 21, 22). A retrospective study in 2021 indicated that the patients' C-peptide level were 0.1 ng/mL (0.012–3.16 ng/mL) (18). In this case, multiple measurements of C-peptide were all <0.09 ng/ml. According to the definition of fulminant type 1 diabetes in Japan, C-peptide is one of the important indicators (21). The diagnostic criteria (28) are as follows: hyperglycemia develops into ketosis or ketoacidosis within 1 week; Blood glucose ≥16.0 mmol/L and HbA1c <8.5% at first diagnosis; Urinary C-peptide <10 μg/d or fasting serum C-peptide <0.3 ng/mL and postprandial C-peptide <0.5 ng/mL. As an important diagnostic indicator of fulminant diabetes, C-peptide should be closely monitored, especially for patients with rapidly elevated blood glucose (22).

Diabetes-Related Autoantibodies

For patients with autoimmune diabetes, diabetes-related autoantibodies, including glutamic acid decarboxylase antibody (GADA), islet-cell antibody (ICA), insulin antibody (IAA), anti-insulinoma antigen-2 antibody (IA-2A), and zinc transporter 8 autoantibody (ZnT8), are usually tested (22). Among patients with anti-PD-1 diabetes, 50–56% were detected at least one diabetes-related autoantibody, and the most prevalent were GADA (1, 4, 5, 9, 11, 16). Chang et al. (20) pointed out that autoantibody detection may not be useful as a biomarker to predict the occurrence of anti-PD-1 related diabetes. Patients could have autoantibody before or after anti-PD-1 treatment, or turn from positive to negative after treatment. However, Gauci et al. (21) found that autoantibodies were associated with the length of time interval between the onset of autoimmune diabetes. GADA-positive patients had a shorter median interval between onsets than negative ones (1, 4, 5, 9, 16, 26, 29). Akturk et al. (16) also pointed out that autoantibody positive patients were more likely to develop DKA (86 vs. 60%; P = 0.02). Therefore, diabetes-related autoantibodies are useful biomarkers and should be detected when necessary.

Pancreatic Exocrine Enzyme and Pancreatic Imaging Examination

20–52% of patients with anti-PD-1 diabetes had elevated lipase or amylase, suggesting inflammation of the pancreas, which may be related to destruction of islet β cells (13, 30). In addition, some scholars observed pancreatic enlargement before the onset of diabetes and progressive pancreatic atrophy afterwards, which may also be used as an indicator for early diagnosis or monitoring (14, 27). However, some scholars reported normal or no significant changes in pancreatic imaging (20). Further studies are needed to understand the relationship among pancreatic exocrine enzyme, imaging and anti-PD-1 diabetes.

Diabetes Related Gene Test

The development of anti-PD-1 diabetes is also related to the susceptibility of genes. Among the patients whose HLA genotypes were tested, 61–76% had genotypes related to ICIs-induced diabetes, much higher than the normal population, and HLA-DR4 was the dominant one (6, 13, 15). HLA-DR4 can be used as a risk predictor of type 1 diabetes induced by anti-PD-1 (2). As HLA genotypes are expensive and time-consuming, their potential as predictors remains to be further studied (15, 18). The author believes that high-risk HLA testing should be examined on patients to be treated with anti-PD-1 to evaluate the risk of diabetes if permission.

The Diagnosis of Anti-PD-1 Diabetes

In this case, the patient had no previous history of diabetes, and the level of blood glucose was normal before treatment. After three cycles of anti-PD-1 therapy, the blood glucose was 30.32 mmol/L, HbA1c was 7.6%, and fasting and postprandial C-peptide were all <0.1 ng/ mL, suggesting rapid destruction of pancreatic β cells. In addition, the patient developed symptoms of diabetic ketoacidosis rapidly after hyperglycemia, so the diagnosis was considered as fulminant type 1 diabetes induced by anti-PD-1 therapy. The patient was also treated with hepatic arterial infusion chemotherapy. According to relevant literature reports, oxaliplatin and 5-fluorouracil may also induce diabetes (23, 3133). However, oxaliplatin and 5-fluorouracil damage islet β cells through damaging the pancreas or causing pancreatic inflammation, which would influent insulin's synthesis and secretion, finally resulting in abnormal blood glucose regulation (3135). In this patient, we adopt hepatic artery infusion chemotherapy (FOLFOX protocol: Oxaliplatin 130 mg/m2, Calcium 48 levofolinate 200 mg/m2, 5-fluorouracil 400 mg/m2) which lasted 48 h so that oxaliplatin and 5-fluorouracil could not reach the pancreas or the dose was very small which may not lead to diabetes. At the same time, the chemotherapy-induced diabetes reported by the relevant literature was all caused by systemic chemotherapy, and there were no reports on hepatic artery infusion chemotherapy. Therefore, the possibility of oxaliplatin and 5-fluorouracil inducing diabetes is basically ruled out.

The Therapy and Prognosis of Anti-PD-1 Diabetes

For the treatment of diabetes induced by anti-PD-1, 71 patients in 56 literatures reviewed by Akturk et al. (16) all required continuous insulin treatment afterwards. Among the 66 patients reported by Chang et al. (20), only one patient stopped insulin treatment 54 days after the onset of diabetes. Autoimmune diabetes caused by anti-PD-1 is irreversible and is associated with massive destruction of islet β cells (3, 18). These patients require long-term insulin to control blood glucose, and continuous insulin treatment is the standard treatment (18, 22, 23). This case required long-term insulin therapy, too.

As an autoimmune disease, whether to use glucocorticoids is controversial. Although some clinicians attempted to use glucocorticoids to salvage islet function, unlike other autoimmune diseases, glucocorticoids therapy had no effect, which may be related to the complete destruction of β cells (14, 36). Chang et al. (20) reviewed the cases of systemic hormone therapy and noted that none of the patients were able to reverse diabetes. Moreover, glucocorticoid treatment may lead to deterioration of blood glucose and insulin resistance in some patients (1, 10, 22). Therefore, hormone therapy is not recommended for the patient and was not used in this case.

Whether to continue anti-PD-1 treatment and when to continue treatment also depends on the severity of the patient's diabetes. According to the criteria of tumor treatment-related adverse events, anti-PD-1-induced diabetes can be divided into five grades according to severity: Grade 1, asymptomatic or mild symptoms and fasting blood glucose >7 mmol/L (126 mg/dL); Grade 2, moderate symptoms and fasting blood glucose >8.9 mmol/L (160 mg/dl); Grade 3, severe but not life-threatening symptoms, requiring hospitalization; Grade 4, life-threatening symptoms requiring emergency intervention; Grade 5, death (11). For grade 1 patients, anti-PD-1 therapy can be continued; Patients with grade 2–3 need to stop anti-PD-1 and receive insulin therapy; Grade 4 patients need to be treated with DKA and monitored (17, 37). By the time patients develop clinical symptoms, 80–95% of pancreatic cells are permanently destroyed, so anti-PD-1 treatment can be resumed when glycemic control is stabilized (17, 38, 39).

While anti-PD-1 diabetes can be life-threatening, patients may also have better anti-tumor effects (1, 36). Judd et al. (40) and Morganna et al. (41) both noted that patients with immune-related adverse reactions caused by anti-PD-1 had longer overall survival, longer progression-free survival and higher survival rate.

Conclusion

As more and more research has confirmed the therapeutic value of anti-PD-1 in malignant tumors, various adverse reactions caused by immunotherapy, such as diabetes, are also increasing. Although the incidence of anti-PD-1 diabetes is low, due to its non-specific symptoms and frequent occurrence of DKA, it may lead to hospitalization or even death, so it must be paid enough attention. In the process of anti-PD-1 treatment, in addition to informing patients of side effects and corresponding symptoms and signs, it is necessary to closely monitor blood glucose and HbA1c before and during treatment, and if necessary, C peptide, diabetes-related autoantibodies, pancreatic exocrine enzymes and pancreatic imaging and even genotypes such as HLA-DR4 can be examined to help diagnosis, in order to identify and treat diabetes earlier and to avoid treatment-related serious adverse events.

Data Availability Statement

The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.

Ethics Statement

Ethical review and approval was not required for the study on human participants in accordance with the local legislation and institutional requirements. The patients/participants provided their written informed consent to participate in this study.

Author Contributions

Z-KZ researched data and wrote the manuscript. Z-GZ revised the manuscript. J-LW, W-XL, T-QW, and M-SC contributed to the treatment of this patient. All authors contributed to the discussion of the article and agree to be accountable for the content of the work. All authors contributed to the article and approved the submitted version.

Funding

This study was funded by Special fund for clinical research of Wu Jieping Foundation (320.6750.2021-02-76) and Sun Yat-sen University Cancer Center Physician Scientist Funding (No. 16zxqk04).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We would like to thank the patient for allowing the publication of this case. We thank the department of liver surgery, Sun Yat-sen University Cancer Center, Guangzhou, China for the full support of laboratory tests.

References

1. Clotman K, Janssens K, Specenier P, Weets I, De Block CEM. Programmed cell death-1 inhibitor-induced type 1 diabetes mellitus. J Clin Endocrinol Metab. (2018) 103:3144–54. doi: 10.1210/jc.2018-00728

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Magis Q, Gaudy-Marqueste C, Basire A, Loundou A, Malissen N, Troin L, et al. Diabetes and blood glucose disorders under anti-PD1. J Immunother. (2018) 41:232–40. doi: 10.1097/CJI.0000000000000218

PubMed Abstract | CrossRef Full Text | Google Scholar

3. De Filette J, Andreescu CE, Cools F, Bravenboer B, Velkeniers B. A systematic review and meta-analysis of endocrine-related adverse events associated with immune checkpoint inhibitors. Hormone Metab Res. (2019) 51:145–56. doi: 10.1055/a-0843-3366

PubMed Abstract | CrossRef Full Text | Google Scholar

4. De Filette JMK, 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. doi: 10.1530/EJE-19-0291

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Abdullah HMA, Elnair R, Khan UI, Omar M, Morey-Vargas OL. Rapid onset type-1 diabetes and diabetic ketoacidosis secondary to nivolumab immunotherapy: a review of existing literature. BMJ Case Rep. (2019) 12:568. doi: 10.1136/bcr-2019-229568

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Dougan M, Pietropaolo M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J Clin Invest. (2020) 130:51–61. doi: 10.1172/JCI131194

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Chen W-X, Li G-X, Hu Z-N, Zhu P, Zhang B-X, Ding Z-Y. Significant response to anti-PD-1 based immunotherapy plus lenvatinib for recurrent intrahepatic cholangiocarcinoma with bone metastasis: a case report and literature review. Medicine. (2019) 98:832. doi: 10.1097/MD.0000000000017832

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Long LZ, Xin L, Hong P, Wei PZ, Ting LJ, Di T, et al. Anti-PD-1 immunotherapy and radiotherapy for stage iv intrahepatic cholangiocarcinoma: a case report. Front Med. (2020) 7:368. doi: 10.3389/fmed.2020.00368

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Cukier P, Santini FC, Scaranti M, Hoff AO. Endocrine side effects of cancer immunotherapy. Endocr Relat Cancer. (2017) 24:331–47. doi: 10.1530/ERC-17-0358

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Tan MH, Iyengar R, Mizokami-Stout K, Yentz S, Maceachern MP, Shen LY, et al. Spectrum of immune checkpoint inhibitors-induced endocrinopathies in cancer patients: a scoping review of case reports. Clin Diabetes Endocrinol. (2019) 5:1. doi: 10.1186/s40842-018-0073-4

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Paschou SA, Stefanaki K, Psaltopoulou T, Liontos M, Koutsoukos K, Zagouri F, et al. How we treat endocrine complications of immune checkpoint inhibitors. ESMO Open. (2021) 6:100011. doi: 10.1016/j.esmoop.2020.100011

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Barroso-Sousa R, Barry WT, Garrido-Castro AC, Hodi FS, Min L, Krop IE, et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. (2018) 4:173–82. doi: 10.1001/jamaoncol.2017.3064

PubMed Abstract | CrossRef Full Text | Google Scholar

13. 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. doi: 10.2337/dbi18-0002

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Marchand L, Thivolet A, Dalle S, Chikh K, Reffet S, Vouillarmet J, et al. Diabetes mellitus induced by PD-1 and PD-L1 inhibitors: description of pancreatic endocrine and exocrine phenotype. Acta Diabetol. (2019) 56:441–8. doi: 10.1007/s00592-018-1234-8

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Youssef N, Noureldein M, Daoud G, Eid AA. Immune checkpoint inhibitors and diabetes: mechanisms and predictors. Diabetes Metab. (2021) 47:101193. doi: 10.1016/j.diabet.2020.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

16. 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. Diabetic Med. (2019) 36:1075–81. doi: 10.1111/dme.14050

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Agrawal L, Bacal A, Jain S, Singh V, Emanuele N, Emanuele M, et al. Immune checkpoint inhibitors and endocrine side effects, a narrative review. Postgrad Med. (2020) 132:206–14. doi: 10.1080/00325481.2019.1709344

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Farina KA, Kane MP. Programmed cell death-1 monoclonal antibody therapy and type 1 diabetes mellitus: a review of the literature. J Pharm Pract. (2021) 34:133–40. doi: 10.1177/0897190019850929

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Zamani MR, Aslani S, Salmaninejad A, Javan MR, Rezaei N. PD-1/PD-L and autoimmunity: a growing relationship. Cell Immunol. (2016) 310:27–41. doi: 10.1016/j.cellimm.2016.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev. (2019) 40:17–65. doi: 10.1210/er.2018-00006

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Gauci ML, Laly P, Vidal-Trecan T, Baroudjian B, Gottlieb J, Madjlessi-Ezra N, et al. Autoimmune diabetes induced by PD-1 inhibitor-retrospective analysis and pathogenesis: a case report and literature review. Cancer Immunol Immunother. (2017) 66:1399–410. doi: 10.1007/s00262-017-2033-8

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Li W, Wang H, Chen B, Zhao S, Zhang X, Jia K, et al. Anti PD-1 monoclonal antibody induced autoimmune diabetes mellitus: a case report and brief review. Transl Lung Cancer Res. (2020) 9:379–88. doi: 10.21037/tlcr.2020.03.05

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Zhang R, Cai XL, Liu L, Han XY, Ji LN. Type 1 diabetes induced by immune checkpoint inhibitors. Chin Med J. (2020) 133:2595–8. doi: 10.1097/CM9.0000000000000972

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Kyriacou A, Melson E, Chen W, Kempegowda P. Is immune checkpoint inhibitor-associated diabetes the same as fulminant type 1 diabetes mellitus? Clin Med. (2020) 20:417–23. doi: 10.7861/clinmed.2020-0054

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Rajasalu T, Brosi H, Schuster C, Spyrantis A, Boehm BO, Chen L, et al. Deficiency in B7-H1 (PD-L1)/PD-1 coinhibition triggers pancreatic beta-cell destruction by insulin-specific, murine CD8 T-cells. Diabetes J Am Diab Assoc. (2010) 59:1966–73. doi: 10.2337/db09-1135

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. (2017) 13:195–207. doi: 10.1038/nrendo.2016.205

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Byun DJ, Braunstein R, Flynn J, Zheng J, Lefkowitz RA, Kanbour S, et al. Immune checkpoint inhibitor-associated diabetes: a single-institution experience. Diabetes Care. (2020) 43:3106–9. doi: 10.2337/dc20-0609

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Toshiaki H, Akihisa I. Fulminant type 1 diabetes: a novel clinical entity requiring special attention by all medical practitioners. Nat Clin Pract Endocrinol Metab. (2007) 3:36–45. doi: 10.1038/ncpendmet0351

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Yuko U, Hibiki U, Shingo M, Kenjiro I, Ken O, Masayuki I, et al. Association of serum anti-GAD antibody and HLA haplotypes with type 1 diabetes mellitus triggered by nivolumab in patients with non-small cell lung cancer. J Thorac Oncol. (2017) 12:41–3. doi: 10.1016/j.jtho.2016.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Friedman CF, Clark V, Raikhel AV, Barz T, Shoushtari AN, Momtaz P, et al. Thinking critically about classifying adverse events: incidence of pancreatitis in patients treated with nivolumab + ipilimumab. J Natl Cancer Inst. (2017) 109:djw260. doi: 10.1093/jnci/djw260

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Nguyen NP, Vos P, Vinh-Hung V, Borok TL, Dutta S, Karlsson U, et al. Altered glucose metabolism during chemoradiation for head and neck cancer. Anticancer Res. (2009) 29:4683–7.

PubMed Abstract | Google Scholar

32. Feng JP, Yuan XL, Li M, Fang J, Xie T, Zhou Y, et al. Secondary diabetes associated with 5-fluorouracil-based chemotherapy regimens in non-diabetic patients with colorectal cancer: results from a single-centre cohort study. Colorectal Dis. (2013) 15:27–33. doi: 10.1111/j.1463-1318.2012.03097.x

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Carly Y, Nassrein H, Terra A. Capecitabine-induced hyperosmolar hyperglycaemic state. BMJ Case Rep. (2021) 14:241109. doi: 10.1136/bcr-2020-241109

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Han G-H, Huang J-X. Hypertriglyceridemia and hyperglycemia induced by capecitabine: a report of two cases and review of the literature. J Oncol Pharm Pract. (2015) 21:380–3. doi: 10.1177/1078155214532508

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Yang J, Jia B, Qiao Y, Chen W, Qi X. Variations of blood glucose in cancer patients during chemotherapy. Niger J Clin Pract. (2016) 19:704–8. doi: 10.4103/1119-3077.187323

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Aleksova J, Lau PKH, Soldatos G, Mcarthur G. Glucocorticoids did not reverse type 1 diabetes mellitus secondary to pembrolizumab in a patient with metastatic melanoma. BMJ Case Reports. (2016) 2016:bcr2016217454. doi: 10.1136/bcr-2016-217454

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the immune-related adverse effects of immune checkpoint inhibitors: a review. JAMA Oncol. (2016) 2:1346. doi: 10.1001/jamaoncol.2016.1051

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S, et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer. (2016) 60:190–209. doi: 10.1016/j.ejca.2016.02.025

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Miyoshi Y, Ogawa O, Oyama Y. Nivolumab, an anti-programmed cell death-1 antibody, induces fulminant type 1 diabetes. Tohoku Univ Med Press. (2016) 239:155–8. doi: 10.1620/tjem.239.155

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Judd J, Zibelman M, Handorf E, O'neill J, Ramamurthy C, Bentota S, et al. Immune-related adverse events as a biomarker in non-melanoma patients treated with programmed cell death 1 inhibitors. Oncologist. (2017) 22:1232–37. doi: 10.1634/theoncologist.2017-0133

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Morganna F-K, Youngchul K, Heather C, Allison R, Geoffrey G, Weber JS, et al. Nivolumab in resected and unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes. Clin Cancer Res. (2016) 22:886–94. doi: 10.1158/1078-0432.CCR-15-1136

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immune checkpoint inhibitors, anti-programmed cell death protein-1, immune-related adverse events, type 1 diabetes, intrahepatic cholangiocarcinoma

Citation: Zheng Z-K, Wang J-L, Li W-X, Wu T-Q, Chen M-S and Zhou Z-G (2022) Anti-programmed Cell Death Protein-1 Therapy in Intrahepatic Cholangiocarcinoma Induced Type 1 Diabetes: A Case Report and Literature Review. Front. Public Health 10:917679. doi: 10.3389/fpubh.2022.917679

Received: 12 April 2022; Accepted: 25 May 2022;
Published: 16 June 2022.

Edited by:

Rishi Raj, Pikeville Medical Center, United States

Reviewed by:

Pinzhu Huang, Beth Israel Deaconess Medical Center and Harvard Medical School, United States
Shang Minjie, Zhejiang Provincial People's Hospital, China
Xiaohui Wang, Hunan Provincial People's Hospital, China

Copyright © 2022 Zheng, Wang, Li, Wu, Chen and Zhou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zhong-Guo Zhou, zhouzhg@sysucc.org.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.