Background
Since the past 10 years, immune checkpoint inhibitors (ICIs), including programmed cell death protein-1 (PD-1) inhibitors, programmed death-ligand 1 (PD-L1) inhibitors, and cytotoxic T cell-associated protein-4 (CTLA-4) inhibitors, have become a remarkable therapeutic improvement for patients with advanced malignancy.[1] Along with their antitumor process, the overactivated immune cells could result in autoimmune damage in various organs including skin, gastrointestinal, pulmonary, cardiac, renal, and endocrine systems, which are called immune-related adverse events (irAEs).[2] ICI-related endocrinopathies are most common. ICI-related thyroid dysfunction (ICI-TD) is the leading subtype and hypophysitis is ranked second; these are followed by ICI-related insulin-deficient diabetes mellitus (DM), primary adrenal insufficiency (PAI), and autoimmune polyendocrine syndrome (APS), which have been described in several case reports.[3] Although most subtypes of endocrinopathies were mild, rare cases of thyrotoxic crisis, myxedema crisis, and adrenal crisis induced by ICI have also been reported. Therefore, comprehensive data of ICI-related endocrinopathies is critical for clinicians. In the present study, we aim to give new insights about ICI-related endocrinopathies based on recent clinical evidence and guidelines.
Underlying Mechanism of ICI-related Endocrinopathies
The detailed mechanism of ICI-related endocrinopathies remains unclear. There is slight difference of T cell modulation among the three kinds of ICIs. CTLA-4 is expressed after T cell activation and it downregulates T cell activation by competitive inhibition of co-stimulation of CD28 with B7 ligands.[4] Also, PD-1 conducts a negative costimulatory function through combination with PD-L1, and thus, it leads to attenuated T cell activation and tumor immune escape.[5] In patients with ICI-TDs, presence of significantly increased CD4+PD1+ T lymphocytes was proved by flow cytometry of thyroid fine-needle aspiration.[6] Furthermore, in rodent models, it was revealed that cytotoxic memory CD4+ T cell was activated by PD-1 antibody and led to anti-PD-1 therapy-related destructive thyroiditis.[7] The pituitary glands at autopsy of six patients with CTLA-4 blockade-induced hypophysitis proved the expression of CTLA-4 and extensive destruction after immune reactions.[8] Another study proved that treatment with anti-CTLA-4 antibody precipitated pituitary cells’ destruction via classic complement pathway.[9] Similar to hypophysitis, a provocative hypothesis of the pathogenesis of ICI-related diabetes was that PD-L1 was highly expressed by pancreatic β cells that were resistant to T cell-mediated apoptosis.[10] Therefore, PD-L1 inhibitors brought about immune response in the β cells surviving for a long term and resulted in rapid-onset fulminant diabetes.[10] However, the mechanism of PAI is still unknown. The different local expression of immune molecules in each affected endocrine gland may determine the varied impairment partly induced by each subtype of ICIs (Table 1).
The mechanism and predisposition of endocrinopathies induced by different subtypes of ICIs
Subtype | Autoimmune mediated? | Mechanistic insights | HLA associated? | Most common induced ICIs | Insights into susceptibility of endocrine AEs |
---|---|---|---|---|---|
Thyroid dysfunction[6,7,11,15,20] | Yes | Increased CD4+PD1+ T lymphocytes in FNA. Activated cytotoxic memory CD4+ T cells in PD-1 inhibitor-induced destructive thyroiditis | No | PD-1 inhibitor | Combination therapy increased the risk of ICI-related thyroiditis |
Hypophysitis[8,9,20] | Yes | CTLA-4 expression in pituitary cells. Activation of classic complement pathway by anti-CTLA-4 antibody during pituitary cell destruction | Probable | CTLA-4 inhibitor | Dose-dependent manner was seen in the incidence of CTLA-4 inhibitor-induced hypophysitis |
DM[10,12,27] | Yes | PD-L1 upregulation on islet β cells | Strongly associated | PD-1 or PD-L1 inhibitor | Upregulation of PD-L1 on pancreatic β cells |
PAI[30] | Yes | Insufficient evidence | No | PD-1 inhibitor | Insufficient evidence |
APS[33] | Yes | Insufficient evidence | Insufficient evidence | PD-1 inhibitor monotherapy in 60.9% cases | Insufficient evidence |
DM: diabetes mellitus; PAI: primary adrenal insufficiency; APS: autoimmune polyendocrine syndrome; FNA: fine-needle aspiration; HLA: human leukocyte antigen; PD-1: programmed cell death protein-1; PD-L1: programmed death-ligand 1; CTLA-4: cytotoxic T cell-associated protein-4; ICIs: immune checkpoint inhibitors.
There is a lack of study on the biopsy of ICI-related endocrinopathies. Aside from the pathology study of hypophysitis mentioned above, a case report of the pathological findings of ICI-related thyroiditis revealed distinguishing features including clusters of necrotic cells and lymphocytes’ infiltration.[11] The injury of endocrine glands by ICIs may provide a natural model of immune damage to endocrine organs, which would help investigate the underlying mechanisms of autoimmune endocrine disease from a novel perspective.
Predisposition of endocrine AES of different subtypes of ICIS
The heterogeneity in different subtypes of ICI-related endocrinopathies is found in the three kinds of immunotherapy (Table 1). Genetic factors showed a predisposition in DM and APS induced by ICIs.[12] Human leukocyte antigen (HLA) DR4 (51.3%) was predominantly seen in patients with ICI-related diabetes,[12] while it was reported that HLA-DR and HLA-DQ were associated with fulminant type 1 diabetes or type 1 diabetes with positive glutamate decarboxylase (GAD) antibody.[13, 14] Immunotherapy may be a contributing factor in the population with predisposing HLA genotype and accelerated disease progression in ICI-related DM. In contrast, no predominance of HLA was discovered in patients with ICI-TDs.[15]
Other predisposing factors of irAEs include higher dose of CTLA-4 blockade, combined therapy of ICIs, and preexisting autoimmune diseases.[16] A multicenter observational study found elevated incidence of irAEs in patients with preexisting systemic autoimmune diseases, while the risk of grade 3/4 irAEs was insignificantly different.[17] A future research direction is to investigate whether patients with preexisting endocrine autoimmune diseases, such as autoimmune thyroid diseases, are predisposed to ICI-related endocrinopathies and safety of immunotherapy in them.
Severity of ICI-related endocrinopathies
ICI-related endocrinopathies show mostly mild presentation based on the Common Terminology Criteria for Adverse Events (CTCAE) criteria,[18] and the incidence of grade 3/4 irAEs in endocrine organs does not exceed 1%.[12] In a retrospective study using the World Health Organization (WHO) adverse drug reaction database, VigiBase, the incidence of ICI-TDs in grade 3/4 was 0.68%, with the affected patients showing major or fatal consequences.[19] To date, there is still a lack of real-world evidence from large populations.
Clinical manifestations of ICI-related endocrinopathies
The incidence and manifestations of each ICI-related endocrinopathy are listed in Table 2.[20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33] ICI-induced APS is even rare. In our previous study, the likelihood of APS in patients on combination therapy of anti-CTLA-4 and anti-PD-1 increased onefold than in patients on PD-1 inhibitors and fourfold than in those on PD-L1 inhibitors.[32]
Clinical manifestations of ICI-related endocrinopathies
Endocrinopathies | Incidence (%) | Gender predominance | Classic manifestations | Onset time after ICI initiation |
---|---|---|---|---|
ICI-TDs[20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33] | 2.60–6.07 | Female predominance | Transient thyrotoxicosis preceding hypothyroidism | Thyrotoxicosis at 5.3 weeks |
Graves’ disease with 50% negative TRAb | ||||
11 cases of thyrotoxic crisis and one case of myxedema were reported | Hypothyroidism at 10.4 weeks | |||
Hypophysitis[20,24, 25, 26] | 3.2–6.4 | Male:female ratio 4:1 | Symptoms included headache and presentations of pituitary hormone dysfunction | Several weeks to 3 months |
Radiologic sign was pituitary stalk enlargement | ||||
Secondary adrenal insufficiency was most common, followed by central hypothyroidism, hypogonadotropic hypogonadism, and GH deficiency. Diabetes insipidus rarely occurs | ||||
DM[16,27,28, 29] | 1.18–2.60 | No gender predominance | Rapid onset of hyperglycemia and progression of insulin deficiency. DKA occurrence rate was 70% | At 1 week to 12 months, median onset |
In most cases at diagnosis, glucose level exceeded 18 mmol/L, with HbA1c less than 8.0% and random C-peptide less than 0.1 ng/mL | time was 3 months | |||
50% cases displayed positive autoimmune diabetic | ||||
antibodies | ||||
CT scan or MR imaging showed pancreatic enlargement followed by progressive pancreatic atrophy | ||||
Slightly elevated amylase levels | ||||
PAI[30, 31] | 0.9 | The proportion of males was 58.1% | Low cortisol level and elevated ACTH level, severe hyponatremia, hyperkalemia, and reduced aldosterone level | Not enough evidence |
Imaging showed bilateral enlargement of adrenal glands | ||||
Positive 21-hydroxylase adrenal cortex antibody was reported | ||||
APS[33] | Not enough evidence | Not enough evidence | More than one of the above diseases co-occurred with manifestations of each disease | Not enough evidence |
ICIs: immune checkpoint inhibitors; ICI-TDs: ICI-related thyroid dysfunction; DM: diabetes mellitus; PAI: primary adrenal insufficiency; APS: autoimmune polyendocrine syndrome; TRAb: thyroid-stimulating hormone receptor antibody; GH: growth hormone; DKA: diabetic ketoacidosis; PD-1: programmed cell death protein-1; PD-L1: programmed death-ligand 1; CTLA-4: cytotoxic T cell-associated protein-4; CT: computed tomography; MR: magnetic resonance
Debating on the treatment of ICI-related endocrinopathies
For cases of all endocrinopathies in CTCAE grade 1, immunotherapy could be continued during the diagnosis and management of the endocrinopathies. In the case of CTCAE grade 2–4, immunotherapy should be discontinued until the endocrinopathy is managed by standard treatment.[12] Importantly, clinicians are confused about the safety of restarting immunotherapy in patients who have recovered from grade 4 ICI-related endocrinopathies. For grade 3/4 hypophysitis and grade 4 ICI-TDs, the decision of restarting immunotherapy after the endocrinopathy is treated or permanently discontinuing ICI therapy is under debate.[2, 34] It was proved that the risk of both initial irAE and other kinds of new-onset irAE was not increased after resuming PD-1 inhibitors in patients who recovered from severe irAEs.[35] Unfortunately, no similar study focusing on grade 4 ICI-related endocrinopathies was conducted to prove the safety of restarting immunotherapy. Study concerning the clinical outcomes of restarting immunotherapy in patients who recovered from grade 3/4 endocrine AEs is of great importance for taking a clinical decision for patients with advanced malignancy.
For the management of insufficiency of endocrine function, replacement therapy is recommended. The need of lifelong replacement varied in endocrine glands and studies. Among ICI-TDs, a possibility of remission was seen in overt hypothyroidism after thyrotoxicosis in ICI-induced silent thyroiditis. However, damage to thyroid function may continue to reach an irreversible situation. since it is not recommended to discontinue ICI in patients with grade 1/2 ICI-TDs. Thus, in nearly half of the cases, de novo hypothyroidism induced by ICIs needed lifelong replacement therapy.[36] In ICI-related hypophysitis, the inflammatory mass of pituitary resolved in most cases, while hormone deficiency persisted.[37] The spontaneous recovery rate of central hypothyroidism and hypogonadotropic hypogonadism was observed to be 30%–60%,[23] while ICI-related central adrenal insufficiency persisted in all cases, which should be managed with lifelong corticosteroid replacement. There is a rapid progression of deficiency of endogenous insulin secretion in ICI-related DM. Due to permanently compromised insulin secretion, lifelong insulin therapy is needed even in patients with preexisting type 2 diabetes.[28] Likewise, lifelong glucocorticoid replacement in ICI-related PAI is urgent, and it should be given promptly to avoid adrenal crisis.[37]
Endocrine damage induced by ICIs on high-dose steroid treatment is under debate. In patients with grade 4 hypophysitis, high-dose steroids (prednisone/ methylprednisolone 1 mg/kg/d) are recommended to relieve acute severe symptoms until the symptoms resolve in an average 1–2 weeks and taper rapidly to physiologic replacement dose.[38] However, a retrospective study revealed that systemic high-dose corticosteroid (prednisone 40–100 mg/d) treatment did not improve the outcome of ipilimumab-related hypophysitis.[39] To the best of our knowledge, no systematic study regarding the efficacy of high-dose glucocorticoids to suppress the autoimmune damage on endocrine organs on the outcome or reversal of grade 4 ICI-related endocrinopathies has proved, along with the possible higher risk of tumor progression in the treatment with high-dose glucocorticoids due to the decreased antitumor efficacy of immunotherapy.[16] Therefore, in case of ICI-related endocrine crisis such as thyrotoxic crisis, myxedema, or adrenal crisis,[38] treatment with high-dose steroids was recommended for the purpose of relieving acute symptoms. Whether short-term treatment with high-dose steroids in endocrine crisis increased the risk of tumor progression calls for real-world evidence.
Impact on prognosis of ICI-related endocrinopathies
Among all ICI-related endocrinopathies, the prognosis of ICI-related PAI was poor. The risk of severe clinical outcomes in ICI-related PAI was over 90%, and the mortality rate was up to 7.3%.[30]
Interestingly, in tumor prognosis, endocrine adverse events are not always linked to adverse outcomes. Kim et al.[40] found a longer OS and progression-free survival (PFS) in 58 patients who developed ICI-TDs with stage IV non-small-cell lung cancer (NSCLC) under PD-1 blockade. Researchers believed that the development of irAEs could partly reflect the antitumor efficacy in patients on ICI regimens, though it is accompanied with a proportion of immortal time bias.[41] However, our data from FAERS database concluded differently. In patients on ICI therapy with adverse events, the immune-related endocrinopathies, including ICI-TDs and other accompanying endocrinopathies, were positively associated with the risk of severe clinical outcomes comprising hospitalization, disabled or life-threatening situations, while they were negatively related to the risk of death.[32] Therefore, irAEs of endocrine organs are critical clues for clinicians to pay attention to. Once the endocrine function alters and endocrinologic irAEs happen, oncologists or endocrinologists ought to start administering replacement therapy or other management in time to avoid serious clinical outcomes.
New insights into the application of ICIs on malignancies of endocrine system
Evidence has accumulated on the application of ICIs in pulmonary combined large cell neuroendocrine carcinoma (LCNEC), anaplastic thyroid cancer (ATC), and advanced adrenocortical carcinoma (ACC),[42, 43, 44, 45] which displayed aggressive behavior and poor prognosis. PD-L1 had an increased proportion of tumoral expression on ATC,[43] and PD-1 inhibitor pembrolizumab was a promising choice of treatment in ATC, based on small studies.[42] Well-designed randomized controlled trials (RCTs) are still in need to provide stronger evidence on immunotherapy in the above malignancies of endocrine systems.
Perspectives
The wide application of ICIs has brought about a novel spectrum of immune-related endocrinopathies in a significant number of patients. Awareness of the heterogeneity in manifestations of ICI-related endocrinopathies is crucial to guide endocrinologists and oncologists. A main dilemma is how to treat our patients properly with ICIs with the purpose of avoiding AEs and benefiting from antitumor effect. Additionally, ICIs-AE might be a model of immune imbalance for the research of autoimmune diseases.
-
Conflict of Interest
All authors declare that they have no conflict of interests.
References
1 Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019; 30: 1321-8.10.1093/annonc/mdz167Suche in Google Scholar
2 Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018; 36: 1714-68.10.1200/JCO.2017.77.6385Suche in Google Scholar
3 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: 1008-19.10.1001/jamaoncol.2019.0393Suche in Google Scholar
4 Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, Peach R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1994; 1: 793-801.10.1016/S1074-7613(94)80021-9Suche in Google Scholar
5 Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017; 355: 1428-33.10.1126/science.aaf1292Suche in Google Scholar PubMed PubMed Central
6 Kotwal A, Gustafson MP, Bornschlegl S, Kottschade L, Delivanis DA, Dietz AB, et al. Immune checkpoint inhibitor-induced thyroiditis is associated with increased intrathyroidal T lymphocyte subpopulations. Thyroid 2020; 30: 1440-50.10.1089/thy.2020.0075Suche in Google Scholar PubMed PubMed Central
7 Yasuda Y, Iwama S, Sugiyama D, Okuji T, Kobayashi T, Ito M, et al. CD4+ T cells are essential for the development of destructive thyroiditis induced by anti-PD-1 antibody in thyroglobulin-immunized mice. Sci Transl Med 2021; 13: eabb7495.10.1126/scitranslmed.abb7495Suche in Google Scholar PubMed
8 Caturegli P, Di Dalmazi G, Lombardi M, Grosso F, Larman HB, Larman T, et al. Hypophysitis secondary to cytotoxic T- lymphocyte associated protein 4 blockade: insights into pathogenesis from an autopsy series. Am J Pathol 2016; 186: 3225-35.10.1016/j.ajpath.2016.08.020Suche in Google Scholar PubMed PubMed Central
9 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: 230ra45.10.1126/scitranslmed.3008002Suche in Google Scholar PubMed
10 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: 727-38.10.1016/j.cmet.2017.01.005Suche in Google Scholar PubMed PubMed Central
11 Angell TE, Min L, Wieczorek TJ, Hodi FS. Unique cytologic features of thyroiditis caused by immune checkpoint inhibitor therapy for malignant melanoma. Genes Dis 2018; 5: 46-8.10.1016/j.gendis.2017.11.002Suche in Google Scholar
12 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: 337-49.10.1007/s11154-020-09618-wSuche in Google Scholar
13 Çekin Y, Özkaya E, Gülkesen H, Akçurin S, Çolak D. Investigation of enterovirus infections, autoimmune factors and HLA genotypes in patients with T1DM. Minerva Endocrinol 2014; 39: 67-74.Suche in Google Scholar
14 Kwak SH, Kim YJ, Chae J, Lee CH, Han B, Kim JI, et al. Association of HLA genotype and fulminant type 1 diabetes in Koreans. Genomics Inform 2015; 13: 126-31.10.5808/GI.2015.13.4.126Suche in Google Scholar
15 Dougan M, Pietropaolo M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J Clin Invest 2020; 130: 51-61.10.1172/JCI131194Suche in Google Scholar
16 Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, MinL. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev 2019; 40: 17-65.10.1210/er.2018-00006Suche in Google Scholar
17 Cortellini A, Buti S, Santini D, Perrone F, Giusti R, Tiseo M, et al. Clinical outcomes of patients with advanced cancer and pre-existing autoimmune diseases treated with anti-programmed death-1 immunotherapy: a real-world transverse study. Oncologist 2019; 24: e327-37.10.1634/theoncologist.2018-0618Suche in Google Scholar
18 Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomized, controlled, phase 2 trial. Lancet Oncol 2015; 16: 908-18.10.1016/S1470-2045(15)00083-2Suche in Google Scholar
19 Bai X, Chen X, Wu X, Huang Y, Zhuang Y, Lin X. Immune checkpoint inhibitor-associated thyroid dysfunction: a disproportionality analysis using the WHO Adverse Drug Reaction Database, VigiBase. Eur J Endocrinol 2020; 182: 1-9.10.1530/EJE-19-0535Suche in Google Scholar PubMed
20 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: 173-82.10.1001/jamaoncol.2017.3064Suche in Google Scholar PubMed PubMed Central
21 Iyer PC, Cabanillas ME, Waguespack SG, Hu MI, Thosani S, Lavis VR, et al. Immune-related thyroiditis with immune checkpoint inhibitors. Thyroid 2018; 28: 1243-51.10.1089/thy.2018.0116Suche in Google Scholar PubMed PubMed Central
22 Brancatella A, Viola N, Brogioni S, Montanelli L, Sardella C, Vitti P, et al. Graves’ disease induced by immune checkpoint inhibitors: a case report and review of literature. Eur Thyroid J 2019; 8: 192-5.10.1159/000501824Suche in Google Scholar PubMed PubMed Central
23 Khan U, Rizvi H, Sano D, Chiu J, Hadid T. Nivolumab induced myxedema crisis. J Immunother Cancer 2017; 5: 13.10.1186/s40425-017-0213-xSuche in Google Scholar PubMed PubMed Central
24 Atkins P, Ur E. Primary and ipilimumab–induced hypophysitis: A single-center case series. Endocr Res 2020; 45: 246-53.10.1080/07435800.2020.1817064Suche in Google Scholar PubMed
25 Caturegli P, Di Dalmazi G, Lombardi M, Grosso F, Larman HB, Larman T, et al. Hypophysitis secondary to cytotoxic T-lymphocyte- associated protein 4 blockade: insights into pathogenesis from an autopsy series. Am J Pathol 2016; 186: 3225–35.10.1016/j.ajpath.2016.08.020Suche in Google Scholar PubMed PubMed Central
26 Amereller F, Deutschbein T, Joshi M, Schopohl J, Schilbach K, Detomas M, et al. Differences between immunotherapy-induced and primary hypophysitis- a multicenter retrospective study. Pituitary 2022; 25: 152–8.10.1007/s11102-021-01182-zSuche in Google Scholar PubMed PubMed Central
27 Liu J, Zhou H, Zhang Y, Fang W, Yang Y, Huang Y, et al. Reporting of immune checkpoint inhibitor therapy- associated diabetes, 2015-2019. Diabetes Care 2020; 43: e79-e80.10.2337/dc20-0459Suche in Google Scholar PubMed PubMed Central
28 Godwin JL, Jaggi S, Sirisena I, Sharda P, Rao AD, Mehra R, et al. Nivolumab-induced autoimmune diabetes mellitus presenting as diabetic ketoacidosis in a patient with metastatic lung cancer. J Immunother Cancer 2017; 5: 40.10.1186/s40425-017-0245-2Suche in Google Scholar PubMed PubMed Central
29 Kong SH, Lee SY, Yang YS, Kim TM, Kwak SH. Anti-programmed cell death 1 therapy triggering diabetic ketoacidosis and fulminant type 1 diabetes. Acta Diabetol 2016; 53: 853-6.10.1007/s00592-016-0872-ySuche in Google Scholar PubMed
30 Grouthier V, Lebrun-Vignes B, Moey M, Johnson DB, Moslehi JJ, Salem JE, et al. Immune checkpoint inhibitor-associated primary adrenal insufficiency: WHO VigiBase Report Analysis. Oncologist 2020; 25: 696-701.10.1634/theoncologist.2019-0555Suche in Google Scholar PubMed PubMed Central
31 Paepegaey AC, Lheure C, Ratour C, Lethielleux G, Clerc J, Bertherat J, et al. Polyendocrinopathy resulted from pembrolizumab in a patient with a malignant melanoma. J Endocrine Soc 2017; 1: 646-9.10.1210/js.2017-00170Suche in Google Scholar PubMed PubMed Central
32 Lu D, Yao J, Yuan G, Gao Y, Zhang J, Guo X. Immune checkpoint inhibitor- associated new-onset thyroid dysfunction: a retrospective analysis using the US FDA Adverse Event Reporting System. Oncologitst 2022; 27: e126-e132.10.1093/oncolo/oyab043Suche in Google Scholar PubMed
33 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: 1527-40.10.1007/s00262-020-02699-1Suche in Google Scholar PubMed
34 Arima H, Iwama S, Inaba H, Ariyasu H, Makita N, Otsuki M, et al. Management of immune-related adverse events in endocrine organs induced by immune checkpoint inhibitors: clinical guidelines of the Japan Endocrine Society. Endocr J 2019; 66: 581-6.10.1507/endocrj.EJ19-0163Suche in Google Scholar PubMed
35 Pollack MH, Betof A, Dearden H, Rapazzo K, Valentine I, Brohl AS, et al. Safety of resuming anti-PD-1 in patients with immune-related adverse events (irAEs) during combined anti-CTLA-4 and anti-PD1 in metastatic melanoma. Ann Oncol 2018; 29: 250-5.10.1093/annonc/mdx642Suche in Google Scholar PubMed PubMed Central
36 Sznol M, Postow MA, Davies MJ, Pavlick AC, Plimack ER, Shaheen M, et al. Endocrine-related adverse events associated with immune checkpoint blockade and expert insights on their management. Cancer Treat Rev 2017; 58: 70-6.10.1016/j.ctrv.2017.06.002Suche in Google Scholar PubMed
37 Scott ES, Long GV, Guminski A, Clifton-Bligh RJ, Menzies AM, Tsang VH. The spectrum, incidence, kinetics and management of endocrinopathies with immune checkpoint inhibitors for metastatic melanoma. Eur J Endocrinol 2018; 178: 175-82.10.1530/EJE-17-0810Suche in Google Scholar PubMed
38 Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, et al. NCCN Guidelines Insights: management of immunotherapy-related toxicities, Version 1.2020. J Natl Compr Canc Netw 2020; 18: 230-41.10.6004/jnccn.2020.0012Suche in Google Scholar PubMed
39 Min L, Hodi FS, Giobbie-Hurder A, Ott PA, Luke JJ, Donahue H, et al. Systemic high-dose corticosteroid treatment does not improve the outcome of ipilimumab-related hypophysitis: a retrospective cohort study. Clin Cancer Res 2015; 21: 749-55.10.1158/1078-0432.CCR-14-2353Suche in Google Scholar PubMed PubMed Central
40 Kim HI, Kim M, Lee SH, Park SY, Kim YN, Kim H, et al. Development of thyroid dysfunction is associated with clinical response to PD-1 blockade treatment in patients with advanced non-small cell lung cancer. Oncoimmunology 2017; 7: e1375642.10.1080/2162402X.2017.1375642Suche in Google Scholar PubMed PubMed Central
41 Street S, Chute D, Strohbehn I, Zhao S, Rengarajan M, Faje A, et al. The positive effect of immune checkpoint inhibitor-induced thyroiditis on overall survival accounting for immortal time bias: a retrospective cohort study of 6596 patients. Ann Oncol 2021;32: 1050-1.10.1016/j.annonc.2021.05.357Suche in Google Scholar PubMed PubMed Central
42 Bible KC, Kebebew E, Brierley J, Brito JP, Cabanillas ME, Clark TJ Jr, et al. 2021 American Thyroid Association Guidelines for management of patients with anaplastic thyroid cancer. Thyroid 2021; 31: 337-86.10.1089/thy.2020.0944Suche in Google Scholar PubMed PubMed Central
43 Adam P, Kircher S, Sbiera I, Koehler VF, Berg E, Knösel T, et al. FGF-receptors and PD-L1 in anaplastic and poorly differentiated thyroid cancer: evaluation of the preclinical rationale. Front Endocrinol 2121; 12: 712107.10.3389/fendo.2021.712107Suche in Google Scholar PubMed PubMed Central
44 Dudnik E, Kareff S, Moskovitz M, Kim C, Liu SV, Lobachov A, et al. Real-world survival outcomes with immune checkpoint inhibitors in large-cell neuroendocrine tumors of lung. J Immunother Cancer 2021; 9: e001999.10.1136/jitc-2020-001999Suche in Google Scholar PubMed PubMed Central
45 Brabo EP, Moraes AB, Neto LV. The role of immune checkpoint inhibitor therapy in advanced adrenocortical carcinoma revisited: review of literature. J Endocrinol Invest 2020; 43: 1531-42.10.1007/s40618-020-01306-5Suche in Google Scholar PubMed
© 2022 Difei Lu, Ying Gao, published by Sciendo
This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.
Artikel in diesem Heft
- Editorial
- Single-nucleotide polymorphisms in medical nutritional weight loss: Challenges and future directions
- Perspective
- Current development of a nonpharmacological intervention approach for mild cognitive impairment patients and a clinical trial in China
- Immune checkpoint inhibitor-related endocrinopathies
- Commentary
- 10.2478/jtim-2022-0008
- Review Article
- Basic pancreatic lesions: Radiologic-pathologic correlation
- Advances in the application of regenerative medicine in prevention of post-endoscopic submucosal dissection for esophageal stenosis
- Inflammatory factors driving atherosclerotic plaque progression new insights
- Original Article
- Prevalence of left atrial septal pouch among patients with embolic stroke of undetermined source or stroke of known etiology: A retrospective study
- Predicting survival for hepatic arterial infusion chemotherapy of unresectable colorectal liver metastases: Radiomics analysis of pretreatment computed tomography
- Dysregulation of miR-144-5p/RNF187 axis contributes to the progression of colorectal cancer
- Letter to Editor
- The first patient with sporadic Huntington’s disease due to a de novo (CAG)n expansion in China
- Hyperchloremic metabolic acidosis post hysteroscopy: A place for balanced solutions?
Artikel in diesem Heft
- Editorial
- Single-nucleotide polymorphisms in medical nutritional weight loss: Challenges and future directions
- Perspective
- Current development of a nonpharmacological intervention approach for mild cognitive impairment patients and a clinical trial in China
- Immune checkpoint inhibitor-related endocrinopathies
- Commentary
- 10.2478/jtim-2022-0008
- Review Article
- Basic pancreatic lesions: Radiologic-pathologic correlation
- Advances in the application of regenerative medicine in prevention of post-endoscopic submucosal dissection for esophageal stenosis
- Inflammatory factors driving atherosclerotic plaque progression new insights
- Original Article
- Prevalence of left atrial septal pouch among patients with embolic stroke of undetermined source or stroke of known etiology: A retrospective study
- Predicting survival for hepatic arterial infusion chemotherapy of unresectable colorectal liver metastases: Radiomics analysis of pretreatment computed tomography
- Dysregulation of miR-144-5p/RNF187 axis contributes to the progression of colorectal cancer
- Letter to Editor
- The first patient with sporadic Huntington’s disease due to a de novo (CAG)n expansion in China
- Hyperchloremic metabolic acidosis post hysteroscopy: A place for balanced solutions?