107
Views
0
CrossRef citations to date
0
Altmetric
Review

The continuum of care of anticancer treatment-induced hypothyroidism in patients with solid non-thyroid tumors: time for an intimate collaboration between oncologists and endocrinologists

ORCID Icon & ORCID Icon
Pages 531-549 | Received 22 Dec 2021, Accepted 20 Jun 2022, Published online: 01 Aug 2022

References

  • Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249.
  • [cited July 2021]. https://canceratlas.cancer.org/the-burden/cancer-survival/
  • [cited July 2021]. https://seer.cancer.gov/csr/1975_2018.cancer_statistics_review_1975-2018/
  • Kalra S, Priya G, Bhattacharya S, et al. Oncocrinology. J Pak Med Assoc. 2020;70(4):757–761.
  • Ganguly S, Naik D, Muskara A, et al. the nexus of endocrine signaling and cancer: how steroid hormones influence genomic stability. Endocrinology. 2021;162(1):bqaa177.
  • Garmendia Madariaga A, Santos Palacios S, Guillén-Grima F, et al. The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab. 2014;99(3):923–931.
  • Biondi B, Cappola AR, Cooper DS. Subclinical hypothyroidism: a review. JAMA. 2019;322(2):153–160.
  • Mateo RCI, Hennessey JV. Thyroxine and treatment of hypothyroidism: seven decades of experience. Endocrine. 2019;66(1):10–17.
  • Lindholm J, Laurberg P. Hypothyroidism and thyroid substitution: historical aspects. J Thyroid Res. 2011;2011:809341.
  • Sap J, Muñoz A, Damm K, et al. The c-erb-A protein is a high-affinity receptor for thyroid hormone. Nature. 1986;324(6098):635–640.
  • Boelaert K, Franklyn JA. Thyroid hormone in health and disease. J Endocrinol. 2005;187(1):1–15.
  • Gauthier BR, Sola-García A, Cáliz-Molina MÁ, et al. Thyroid hormones in diabetes, cancer, and aging. Aging Cell. 2020;19(11):e13260.
  • Davis PJ, Leonard JL, Lin HY, et al. Molecular basis of nongenomic actions of thyroid hormone. Vitam Horm. 2018;106:67–96.
  • Liu YC, Yeh CT, Lin KH. Molecular functions of thyroid hormone signaling in regulation of cancer progression and anti-apoptosis. Int J Mol Sci. 2019;20(20):4986.
  • Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions. Endocr Rev. 2010;31(2):139–170.
  • Hercbergs A. The thyroid gland as an intrinsic biologic response-modifier in advanced neoplasia–a novel paradigm. Vivo. 1996;10(2):245–247.
  • Goemann IM, Romitti M, Meyer ELS, et al. Role of thyroid hormones in the neoplastic process: an overview. Endocr Relat Cancer. 2017;24(11):R367–R385.
  • Krashin E, Piekiełko-Witkowska A, Ellis M, et al. Thyroid hormones and cancer: a comprehensive review of preclinical and clinical studies. Front Endocrinol (Lausanne). 2019;10:59. DOI:10.3389/fendo.2019.00059.
  • Moriggi G, Verga Falzacappa C, Mangialardo C, et al. Thyroid hormones (T3 and T4): dual effect on human cancer cell proliferation. Anticancer Res. 2011;31(1):89–96.
  • Persani L, Brabant G, Dattani M, et al. 2018 European Thyroid Association (ETA) guidelines on the diagnosis and management of central hypothyroidism. Eur Thyroid J. 2018;7(5):225–237.
  • Salvatore D, Davies TF, Sohlumberger MJ, et al. Chapter 11. Thyroid physiology and diagnostic evaluation of patients with thyroid disorders. In: Melmed S, Koenig R, Rosen C, et al., editors. Williams textbook of endocrinology. 12th Philadelphia: Elsevier Saunders; 2011. p. 261–327.
  • Cooper DS, Landenson PW. Chapter 7. The thyroid gland. In: Gardner D, Greenspan’s SD, editors. Basic and clinical endocrinology. 9th ed. New York United States: The Mc Graw Hill Companies, Inc; 2011. p. 163–226.
  • Burch HB. Drug effects on the thyroid. N Engl J Med. 2019;381(8):749–761.
  • Abualia M, Amarin JZ, Zayed AA. Drug effects on the thyroid. N Engl J Med. 2019;381(20):1979–1980.
  • Hamnvik OP, Larsen PR, Marqusee E. Thyroid dysfunction from antineoplastic agents. J Natl Cancer Inst. 2011;103(21):1572–1587.
  • Carter Y, Sippel RS, Chen H. Hypothyroidism after a cancer diagnosis: etiology, diagnosis, complications, and management. Oncologist. 2014;19(1):34–43. .
  • Bhattacharya S, Goyal A, Kaur P, et al. Anticancer drug-induced thyroid dysfunction. Eur Endocrinol. 2020;16(1): 32–39. .
  • Torino F, Barnabei A, Baldelli R, et al. (July 18th, 2012). Thyroid function abnormalities in patients receiving anticancer agents, thyroid hormone, Neeraj Kumar Agrawal, IntechOpen. Accessed July 21, 2021 doi: 10.5772/50375.
  • Torino F, Barnabei A, Paragliola R, et al. Thyroid dysfunction as an unintended side effect of anticancer drugs. Thyroid. 2013;23(11): 1345–1366. .
  • Reiners C, Drozd V, Yamashita S. Hypothyroidism after radiation exposure: brief narrative review. J Neural Transm (Vienna). 2020;127(11):1455–1466.
  • Baskar R, Lee KA, Yeo R, et al. Cancer and radiation therapy: current advances and future directions. Int J Med Sci. 2012;9(3):193–199.
  • Chalan P, Di Dalmazi G, Pani F, et al. Thyroid dysfunctions secondary to cancer immunotherapy. J Endocrinol Invest. 2018;41(6): 625–638. .
  • Deligiorgi MV, Panayiotidis MI, Trafalis DT. Endocrine adverse events related with immune checkpoint inhibitors: an update for clinicians. Immunotherapy. 2020;12(7):481–510.
  • Gabora K, Piciu A, Bădulescu IC, et al. Current evidence on thyroid related adverse events in patients treated with protein tyrosine kinase inhibitors. Drug Metab Rev. 2019;51(4):562–569.
  • Thomson RJ, Moshirfar M, Ronquillo Y Tyrosine Kinase Inhibitors. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021 Jan. cited 2021 May 4; Accessed July 20, 2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK563322/
  • Mouhayar E, Durand JB, Cortes J. Cardiovascular toxicity of tyrosine kinase inhibitors. Expert Opin Drug Saf. 2013;12(5):687–696.
  • Jonklaas J, Bianco AC, Bauer AJ, et al. American thyroid association task force on thyroid hormone replacement. Guidelines for the treatment of hypothyroidism: prepared by the American thyroid association task force on thyroid hormone replacement. Thyroid. 2014;24(12):1670–1751.
  • Garber JR, Cobin RH, Gharib H, et al. American Association of Clinical Endocrinologists and American Thyroid Association taskforce on hypothyroidism in adults. clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Endocr Pract. 2012;18(6):988–1028.
  • Piras C, Pibiri M, Leoni VP, et al. Analysis of metabolomics profile in hypothyroid patients before and after thyroid hormone replacement. J Endocrinol Invest. 2021;44(6):1309–1319.
  • Köhrle J. The colorful diversity of thyroid hormone metabolites. Eur Thyroid J. 2019;8(3):115–129.
  • Davis PJ, Tang HY, Hercbergs A, et al. Bioactivity of thyroid hormone analogs at cancer cells. Front Endocrinol (Lausanne). 2018;9:739.
  • Louzada RA, Carvalho DP. Similarities and differences in the peripheral actions of thyroid hormones and their metabolites. Front Endocrinol (Lausanne). 2018;9:394.
  • Giammanco M, Di Liegro CM, Schiera G, et al. Genomic and non-genomic mechanisms of action of thyroid hormones and their catabolite 3,5-Diiodo-l-Thyronine in mammals. Int J Mol Sci. 2020;21(11):4140.
  • Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, et al. Hypothalamus-pituitary-thyroid axis. Compr Physiol. 2016;6(3):1387–1428.
  • Davis PJ, Mousa SA, Lin HY. Nongenomic actions of thyroid hormone: the integrin component. Physiol Rev. 2021;101(1):319–352.
  • Flamant F, Cheng SY, Hollenberg AN, et al. Thyroid hormone signaling pathways: time for a more precise nomenclature. Endocrinology. 2017;158(7):2052–2057.
  • Hallen A, Cooper AJ, Jamie JF, et al. Mammalian forebrain ketimine reductase identified as μ-crystallin; potential regulation by thyroid hormones. J Neurochem. 2011;118(3):379–387.
  • Pappas L, Xu XL, Abramson DH, et al. Genomic instability and proliferation/survival pathways in RB1-deficient malignancies. Adv Biol Regul. 2017;64:20–32.
  • Suhane S, Ramanujan VK. Thyroid hormone differentially modulates Warburg phenotype in breast cancer cells. Biochem Biophys Res Commun. 2011;414(1):73–78.
  • Liao CH, Yeh SC, Huang YH, et al. Positive regulation of spondin 2 by thyroid hormone is associated with cell migration and invasion. Endocr Relat Cancer. 2010;17(1):99–111.
  • Kowalik MA, Puliga E, Cabras L, et al. Thyroid hormone inhibits hepatocellular carcinoma progression via induction of differentiation and metabolic reprogramming. J Hepatol. 2020;72(6):1159–1169.
  • De Luca R, Davis PJ, Lin HY, et al. Thyroid hormones interaction with immune response, inflammation and non-thyroidal illness syndrome. Front Cell Dev Biol. 2021;8:614030.
  • Montesinos MDM, Pellizas CG. Thyroid hormone action on innate immunity. Front Endocrinol (Lausanne). 2019;10:350.
  • Perrotta C, Buldorini M, Assi E, et al. The thyroid hormone triiodothyronine controls macrophage maturation and functions: protective role during inflammation. Am J Pathol. 2014;184(1):230–247.
  • Szabó J, Fóris G, Mezösi E, et al. Parameters of respiratory burst and arachidonic acid metabolism in polymorphonuclear granulocytes from patients with various thyroid diseases. Exp Clin Endocrinol Diabetes. 1996;104(2):172–176.
  • Provinciali M, Fabris N. Modulation of lymphoid cell sensitivity to interferon by thyroid hormones. J Endocrinol Invest. 1990;13(2):187–191.
  • Schmohl KA, Müller AM, Wechselberger A, et al. Thyroid hormones and tetrac: new regulators of tumour stroma formation via integrin αvβ3. Endocr Relat Cancer. 2015;22(6):941–952.
  • Zhou L, Chen J, Tao CJ, et al. Research progress of radiation-induced hypothyroidism in head and neck cancer. J Cancer. 2021;12(2): 451–459. .
  • Lin Z, Yang Z, He B, et al. Pattern of radiation-induced thyroid gland changes in nasopharyngeal carcinoma patients in 48 months after radiotherapy. PLoS One. 2018;13(7):e0200310.
  • Boomsma MJ, Bijl HP, Langendijk JA. Radiation-induced hypothyroidism in head and neck cancer patients: a systematic review. Radiother Oncol. 2011;99(1):1–5.
  • Albi E, Cataldi S, Lazzarini A, et al. Radiation and thyroid cancer. Int J Mol Sci. 2017;18(5):911.
  • Rosenthal DI, Chambers MS, Fuller CD, et al. Beam path toxicities to non-target structures during intensity-modulated radiation therapy for head and neck cancer. Int J Radiat Oncol Biol Phys. 2008;72(3):747–755.
  • Kamal M, Peeler CR, Yepes P, et al. Radiation-induced hypothyroidism after radical intensity modulated radiation therapy for oropharyngeal carcinoma. Adv Radiat Oncol. 2020;5(1):111–119.
  • Diaz R, Jaboin JJ, Morales-Paliza M, et al. Hypothyroidism as a consequence of intensity-modulated radiotherapy with concurrent taxane-based chemotherapy for locally advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2010;77(2):468–476.
  • Vogelius IR, Bentzen SM, Maraldo MV, et al. Risk factors for radiation-induced hypothyroidism: a literature-based meta-analysis. Cancer. 2011;117(23):5250–5260.
  • Alba JR, Basterra J, Ferrer JC, et al. Hypothyroidism in patients treated with radiotherapy for head and neck carcinoma: standardised long-term follow-up study. J Laryngol Otol. 2016;130(5):478–481.
  • Luo R, Vwc W, He B, et al. Development of a normal tissue complication probability (NTCP) model for radiation-induced hypothyroidism in nasopharyngeal carcinoma patients. BMC Cancer. 2018;18(1):575.
  • Brodin NP, Kabarriti R, Garg MK, et al. Systematic review of normal tissue complication models relevant to standard fractionation radiation therapy of the head and neck region published after the QUANTEC reports. Int J Radiat Oncol Biol Phys. 2018;100(2):391–407.
  • Nowicka Z, Tomasik B, Papis-Ubych A, et al. Radiation-Induced hypothyroidism in patients with oropharyngeal cancer treated with IMRT: independent and external validation of five normal tissue complication probability models. Cancers (Basel). 2020;12(9):2716.
  • Yeung SC, Chiu AC, Vassilopoulou-Sellin R, et al. The endocrine effects of nonhormonal antineoplastic therapy. Endocr Rev. 1998;19(2):144–172.
  • Stuart NS, Woodroffe CM, Grundy R, et al. Long-term toxicity of chemotherapy for testicular cancer–the cost of cure. Br J Cancer. 1990;61(3):479–484.
  • Ogilvy-Stuart AL, Shalet SM, Gattamaneni HR. Thyroid function after treatment of brain tumors in children. J Pediatr. 1991;119(5):733–737.
  • Massart C, Le Tellier C, Lucas C, et al. Effects of cisplatin on human thyrocytes in monolayer or follicle culture. J Mol Endocrinol. 1992;8(3):243–248.
  • Zatelli MC, Gentilin E, Daffara F, et al. Therapeutic concentrations of mitotane (o,p’-DDD) inhibit thyrotroph cell viability and TSH expression and secretion in a mouse cell line model. Endocrinology. 2010;151(6):2453–2461.
  • Russo M, Scollo C, Pellegriti G, et al. Mitotane treatment in patients with adrenocortical cancer causes central hypothyroidism. Clin Endocrinol (Oxf). 2016;84(4):614–619.
  • Basile V, Puglisi S, Calabrese A, et al. Unwanted hormonal and metabolic effects of postoperative adjuvant mitotane treatment for adrenocortical cancer. Cancers (Basel). 2020;12(9):2615.
  • Aricò E, Castiello L, Capone I, et al. Type I interferons and cancer: an evolving story demanding novel clinical applications. Cancers (Basel). 2019;11(12):1943.
  • Fenton SE, Saleiro D, Platanias LC. Type I and II interferons in the anti-tumor immune response. Cancers (Basel). 2021;13(5):1037.
  • Palacios-Álvarez I, Román-Curto C, Mir-Bonafé JM, et al. Autoimmune response as a side effect of treatment with interferon-α in melanoma: does this have prognostic implications? Int J Dermatol. 2015;54(3):e91–93.
  • Caraccio N, Giannini R, Cuccato S, et al. Type I interferons modulate the expression of thyroid peroxidase, sodium/iodide symporter, and thyroglobulin genes in primary human thyrocyte cultures. J Clin Endocrinol Metab. 2005;90(2):1156–1162.
  • Rizzo LFL, Mana DL, Serra HA. Drug-induced hypothyroidism. Medicina (B Aires). 2017;77(5):394–404.
  • Marabondo S, Kaufman HL. High-dose interleukin-2 (IL-2) for the treatment of melanoma: safety considerations and future directions. Expert Opin Drug Saf. 2017;16(12):1347–1357.
  • Dutcher JP, Schwartzentruber DJ, Kaufman HL, et al. High dose interleukin-2 (Aldesleukin) - expert consensus on best management practices-2014. J Immunother Cancer. 2014;2(1):26.
  • Clark JI, Curti B, Davis EJ, et al. Long-term progression-free survival of patients with metastatic melanoma or renal cell carcinoma following high-dose interleukin-2. J Investig Med. 2021;69(4):888–892.
  • Mortara L, Balza E, Bruno A, et al. Anti-cancer therapies employing IL-2 cytokine tumor targeting: contribution of innate, adaptive and immunosuppressive cells in the anti-tumor efficacy. Front Immunol. 2018;9:2905.
  • Buchbinder EI, Dutcher JP, Daniels GA, et al. Therapy with high-dose Interleukin-2 (HD IL-2) in metastatic melanoma and renal cell carcinoma following PD1 or PDL1 inhibition. J Immunother Cancer. 2019;7(1):49.
  • Yen WC, Corpuz MR, Prudente RY, et al. A selective retinoid X receptor agonist bexarotene (Targretin) prevents and overcomes acquired paclitaxel (Taxol) resistance in human non-small cell lung cancer. Clin Cancer Res. 2004;10(24):8656–8664.
  • Tooker P, Yen WC, Ng SC, et al. Bexarotene (LGD1069, Targretin), a selective retinoid X receptor agonist, prevents and reverses gemcitabine resistance in NSCLC cells by modulating gene amplification. Cancer Res. 2007;67(9):4425–4433.
  • Khuri FR, Rigas JR, Figlin RA, et al. Multi-institutional phase I/II trial of oral bexarotene in combination with cisplatin and vinorelbine in previously untreated patients with advanced non-small-cell lung cancer. J Clin Oncol. 2001;19(10):2626–2637.
  • Edelman MJ, Smith R, Hausner P, et al. Phase II trial of the novel retinoid, bexarotene, and gemcitabine plus carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2005;23(24):5774–5778. Erratum in: J Clin Oncol. 2005 Nov 1;23(31):8142.
  • Dragnev KH, Ma T, Cyrus J, et al. Bexarotene plus erlotinib suppress lung carcinogenesis independent of KRAS mutations in two clinical trials and transgenic models. Cancer Prev Res (Phila). 2011;4(6):818–828.
  • Padda SK, Chhatwani L, Zhou L, et al. Phase I and pharmacokinetic study of bexarotene in combination with gefitinib in the third-line treatment of non-small-cell lung cancer: brief report. Anticancer Drugs. 2013;24(7):731–735.
  • Dragnev KH, Whyman JD, Hahn CK, et al. A phase I/II study of bexarotene with carboplatin and weekly paclitaxel for the treatment of patients with advanced non-small cell lung cancer. J Thorac Dis. 2018;10(9):5531–5537.
  • Rodon J, Jacobs CD, Chu Q, et al. A phase I pharmacokinetic study of bexarotene with paclitaxel and carboplatin in patients with advanced non-small cell lung cancer (NSCLC). Cancer Chemother Pharmacol. 2012;69(3):825–834.
  • Makita N, Manaka K, Sato J, et al. Bexarotene-induced hypothyroidism: characteristics and therapeutic strategies. Clin Endocrinol (Oxf). 2019;91(1):195–200.
  • Scarisbrick JJ, Morris S, Azurdia R, et al. U.K. consensus statement on safe clinical prescribing of bexarotene for patients with cutaneous T-cell lymphoma. Br J Dermatol. 2013;168(1):192–200.
  • Assaf C, Bagot M, Dummer R, et al. Minimizing adverse side-effects of oral bexarotene in cutaneous T-cell lymphoma: an expert opinion. Br J Dermatol. 2006;155(2):261–266.
  • Sherman SI. Etiology, diagnosis, and treatment recommendations for central hypothyroidism associated with bexarotene therapy for cutaneous T-cell lymphoma. Clin Lymphoma. 2003;3(4):249–252.
  • Upadhaya S, Neftelinov ST, Hodge J, et al. Challenges and opportunities in the PD1/PDL1 inhibitor clinical trial landscape [published online ahead of print, 2022 Feb 10]. Nat Rev Drug Discov. 2022. DOI:10.1038/d41573-022-00030-4
  • Raschi E, Mazzarella A, Antonazzo IC, et al. Toxicities with immune checkpoint inhibitors: emerging priorities from disproportionality analysis of the FDA adverse event reporting system. Target Oncol. 2019;14(2):205–221.
  • Deligiorgi MV, Panayiotidis MI, Trafalis DT. Endocrine adverse events related with immune checkpoint inhibitors: an update for clinicians. Immunotherapy. 2020;12(7):481–510.
  • Baxi S, Yang A, Gennarelli RL, et al. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ. 2018;360:k793.
  • Quandt Z, Trupin L, Evans M, et al. Finding the needles in the haystack: harnessing the electronic health record to find thyroid immune related adverse events. J Endocr Soc. 2020;4(Supplement_1):SAT–418.
  • Almutairi AR, McBride A, Slack M, et al. Potential immune-related adverse events associated with monotherapy and combination therapy of ipilimumab, nivolumab, and pembrolizumab for advanced melanoma: a systematic review and meta-analysis. Front Oncol. 2020;10:91.
  • Chang CY, Park H, Malone DC, et al. Immune checkpoint inhibitors and immune-related adverse events in patients with advanced melanoma: a systematic review and network meta-analysis. JAMA Network Open. 2020;3(3):e201611.
  • Xu H, Tan P, Zheng X, et al. Immune-related adverse events following administration of anti-cytotoxic T-lymphocyte-associated protein-4 drugs: a comprehensive systematic review and meta-analysis. Drug Des Devel Ther. 2019;13:2215–2234.
  • de Filette J, Andreescu CE, Cools F, et al. A systematic review and meta-analysis of endocrine-related adverse events associated with immune checkpoint inhibitors. Horm Metab Res. 2019;51(3):145–156.
  • Barroso-Sousa R, Barry WT, Garrido-Castro AC, 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(2):173–182.
  • Da L, Teng Y, Wang N, et al. Organ-specific immune-related adverse events associated with immune checkpoint inhibitor monotherapy versus combination therapy in cancer: a meta-analysis of randomized controlled trials. Front Pharmacol. 2020;10:1671. DOI:10.3389/fphar.2019.01671. .
  • Stelmachowska-Banaś M, Czajka-Oraniec I. Management of endocrine immune-related adverse events of immune checkpoint inhibitors: an updated review. Endocr Connect. 2020;9(10):R207–R228.
  • Muir CA, Menzies AM, Clifton-Bligh R, et al. Thyroid toxicity following immune checkpoint inhibitor treatment in advanced cancer. Thyroid. 2020;30(10):1458–1469.
  • Tan MH, Iyengar R, Mizokami-Stout K, et al. Spectrum of immune checkpoint inhibitors-induced endocrinopathies in cancer patients: a scoping review of case reports. Clin Diabetes Endocrinol. 2019;5:1.
  • Ferrari SM, Fallahi P, Elia G, et al. Autoimmune endocrine dysfunctions associated with cancer immunotherapies. Int J Mol Sci. 2019;20(10):2560.
  • Cukier P, Santini FC, Scaranti M, et al. Endocrine side effects of cancer immunotherapy. Endocr Relat Cancer. 2017;24(12):T331–T347.
  • Corsello SM, Barnabei A, Marchetti P, et al. Endocrine side effects induced by immune checkpoint inhibitors. J Clin Endocrinol Metab. 2013;98(4):1361–1375.
  • Zhai Y, Ye X, Hu F, et al. Endocrine toxicity of immune checkpoint inhibitors: a real-world study leveraging US Food and Drug Administration adverse events reporting system. J Immunother Cancer. 2019;7(1):286.
  • Bai X, Chen X, Wu X, et al. Immune checkpoint inhibitor-associated thyroid dysfunction: a disproportionality analysis using the WHO adverse drug reaction database, vigiBase. Eur J Endocrinol. 2020;182(1):1–9.
  • Orlov S, Salari F, Kashat L, et al. Induction of painless thyroiditis in patients receiving programmed death 1 receptor immunotherapy for metastatic malignancies. J Clin Endocrinol Metab. 2015;100(5):1738–1741.
  • Young A, Quandt Z, Bluestone JA. The balancing act between cancer immunity and autoimmunity in response to immunotherapy. Cancer Immunol Res. 2018;6(12):1445–1452.
  • Angell TE, Min L, Wieczorek TJ, et al. Unique cytologic features of thyroiditis caused by immune checkpoint inhibitor therapy for malignant melanoma. Genes Dis. 2018;5(1):46–48.
  • Gowen MF, Giles KM, Simpson D, et al. Baseline antibody profiles predict toxicity in melanoma patients treated with immune checkpoint inhibitors. J Transl Med. 2018;16(1):82.
  • Chang LS, Barroso-Sousa R, Tolaney SM, et al. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev. 2019;40(1):17–65.
  • Kotwal A, Gustafson MP, Bornschlegl S, et al. Immune checkpoint inhibitor-induced thyroiditis is associated with increased intrathyroidal T lymphocyte subpopulations. Thyroid. 2020;30(10):1440–1450.
  • Olsson-Brown A, Lord R, Sacco J, et al. Two distinct clinical patterns of checkpoint inhibitor-induced thyroid dysfunction. Endocr Connect. 2020;9(4):318–325.
  • Kurimoto C, Inaba H, Ariyasu H, et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune-checkpoint inhibitors. Cancer Sci. 2020;111(5):1468–1477.
  • Kobayashi T, Iwama S, Yasuda Y, et al. Patients with antithyroid antibodies are prone to develop destructive thyroiditis by nivolumab: a prospective study. J Endocr Soc. 2018;2(3):241–251.
  • Delivanis DA, Gustafson MP, Bornschlegl S, et al. Pembrolizumab-Induced thyroiditis: comprehensive clinical review and insights into underlying involved mechanisms. J Clin Endocrinol Metab. 2017;102(8):2770–2780.
  • Thapi S, Leiter A, Galsky M, et al. Recovery from secondary adrenal insufficiency in a patient with immune checkpoint inhibitor therapy induced hypophysitis. J Immunother Cancer. 2019;7(1):248.
  • Ramos-Levi AM, Rogado J, Sanchez-Torres JM, et al. Nivolumab-induced thyroid dysfunction in patients with lung cancer. Endocrinol Diabetes Nutr (Engl Ed). 2019;66(1):26–34.
  • Elia G, Ferrari SM, Galdiero MR, et al. New insight in endocrine-related adverse events associated to immune checkpoint blockade. Best Pract Res Clin Endocrinol Metab. 2020;34(1):101370.
  • Mazarico I, Capel I, Giménez-Palop O, et al. Low frequency of positive antithyroid antibodies is observed in patients with thyroid dysfunction related to immune check point inhibitors. J Endocrinol Invest. 2019;42(12):1443–1450.
  • Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract. 2014;2014:357027.
  • Makita N, Iiri T. Tyrosine kinase inhibitor-induced thyroid disorders: a review and hypothesis. Thyroid. 2013;23(2):151–159.
  • Jiao Q, Bi L, Ren Y, et al. Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol Cancer. 2018;17(1):36.
  • de Groot JW, Zonnenberg BA, Plukker JT, et al. Imatinib induces hypothyroidism in patients receiving levothyroxine. Clin Pharmacol Ther. 2005;78(4):433–438.
  • Pani F, Atzori F, Baghino G, et al. Thyroid dysfunction in patients with metastatic carcinoma treated with sunitinib: is thyroid autoimmunity involved? Thyroid. 2015;25(11):1255–1261.
  • Ahmadieh H, Salti I. Tyrosine kinase inhibitors induced thyroid dysfunction: a review of its incidence, pathophysiology, clinical relevance, and treatment. Biomed Res Int. 2013;2013:725410.
  • Bilen MA, Patel A, Hess KR, et al. Association between new-onset hypothyroidism and clinical response in patients treated with tyrosine kinase inhibitor therapy in phase I clinical trials. Cancer Chemother Pharmacol. 2016;78(1):167–171.
  • Si X, Zhang L, Wang H, et al. Management of anlotinib-related adverse events in patients with advanced non-small cell lung cancer: experiences in ALTER-0303. Thorac Cancer. 2019;10(3):551–556.
  • Funakoshi T, Shimada YJ. Risk of hypothyroidism in patients with cancer treated with sunitinib: a systematic review and meta-analysis. Acta Oncol. 2013;52(4):691–702.
  • Lechner MG, Vyas CM, Hamnvik OR, et al. Hypothyroidism during tyrosine kinase inhibitor therapy is associated with longer survival in patients with advanced nonthyroidal cancers. Thyroid. 2018;28(4):445–453.
  • Wolter P, McCann L, Sternberg CN, et al. Incidence of thyroid dysfunction in renal cell carcinoma (RCC) patients treated with pazopanib in prospective clinical trials. J Clin Oncol. 2011;29(15):4633.
  • Drui D, Illouz F, Do Cao C, et al. Expert opinion on thyroid complications of new anti-cancer therapies: tyrosine kinase inhibitors. Ann Endocrinol (Paris). 2018;79(5):569–573.
  • DeGroot LJ. The non-thyroidal illness syndrome. Feingold KR, Anawalt B, Boyce A, et al., Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000. Updated 2015 Feb 1;Accessed.2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK285570/
  • Keestra S, Högqvist Tabor V, Alvergne A, et al. Reinterpreting patterns of variation in human thyroid function: an evolutionary ecology perspective. Evol Med Public Health. 2020;9(1):93–112.
  • Common Terminology Criteria for Adverse Events (CTCAE) v5.0. Accessed June 2021. Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm
  • Vasileiou M, Gilbert J, Fishburn S, et al. Guideline committee. Thyroid disease assessment and management: summary of NICE guidance. BMJ. 2020;368:m41. DOI:10.1136/bmj.m41. Erratum in: BMJ 2020;368:m437.
  • Calissendorff J, Falhammar H. To treat or not to treat subclinical hypothyroidism, what is the evidence? Medicina (Kaunas). 2020;56(1):40.
  • Benvenga S. L-T4 therapy in the presence of pharmacological interferents. Front Endocrinol (Lausanne). 2020;11:607446.
  • Jonklaas J, Bianco AC, Cappola AR, et al. Evidence-based use of levothyroxine/liothyronine combinations in treating hypothyroidism: a consensus document. Thyroid. 2021;31(2):156–182.
  • Bayat Mokhtari R, Homayouni TS, Baluch N, et al. Combination therapy in combating cancer. Oncotarget. 2017;8(23):38022–38043.
  • Zaheer J, Kim H, Lee YJ, et al. Combination radioimmunotherapy strategies for solid tumors. Int J Mol Sci. 2019;20(22):5579.
  • Kimbara S, Fujiwara Y, Iwama S, et al. Association of antithyroglobulin antibodies with the development of thyroid dysfunction induced by nivolumab. Cancer Sci. 2018;109(11):3583–3590.
  • Funazo TY, Nomizo T, Ozasa H, et al. Clinical impact of low serum free T4 in patients with non-small cell lung cancer treated with nivolumab. Sci Rep. 2019;9(1):17085.
  • Champiat S, Lambotte O, Barreau E, et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol. 2016;27(4):559–574.
  • Antonelli A, Elia G, Ragusa F, et al. The stability of TSH, and thyroid hormones, in patients treated with tablet, or liquid levo-thyroxine. Front Endocrinol (Lausanne). 2021;12:633587.
  • Cornelli U, Belcaro G, Recchia M, et al. Levothyroxine and lung cancer in females: the importance of oxidative stress. Reprod Biol Endocrinol. 2013;11:75.
  • Sarosiek K, Gandhi AV, Saxena S, et al. Hypothyroidism in pancreatic cancer: role of exogenous thyroid hormone in tumor invasion-preliminary observations. J Thyroid Res. 2016;2016:2454989.
  • Boursi B, Haynes K, Mamtani R, et al. Thyroid dysfunction, thyroid hormone replacement and colorectal cancer risk. J Natl Cancer Inst. 2015;107(6):djv084.
  • Hercbergs A. Clinical implications and impact of discovery of the thyroid hormone receptor on integrin αvβ3-A review. Front Endocrinol (Lausanne). 2019;10:565.
  • Osorio JC, Ni A, Chaft JE, et al. Antibody-mediated thyroid dysfunction during T-cell checkpoint blockade in patients with non-small-cell lung cancer. Ann Oncol. 2017;28(3):583–589.
  • Thuillier P, Joly C, Alavi Z, et al. Thyroid dysfunction induced by immune checkpoint inhibitors is associated with a better progression-free survival and overall survival in non-small cell lung cancer: an original cohort study. Cancer Immunol Immunother. 2021;70(7):2023–2033.
  • Kim HI, Kim M, Lee SH, 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(1):e1375642.
  • Campredon P, Mouly C, Lusque A, et al. Incidence of thyroid dysfunctions during treatment with nivolumab for non-small cell lung cancer: retrospective study of 105 patients. Presse Med. 2019;48(4):e199–e207.
  • Sakakida T, Ishikawa T, Uchino J, et al. Clinical features of immune-related thyroid dysfunction and its association with outcomes in patients with advanced malignancies treated by PD-1 blockade. Oncol Lett. 2019;18(2):2140–2147.
  • Freeman-Keller M, Kim Y, Cronin H, et al. Nivolumab in resected and unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes. Clin Cancer Res. 2016;22(4):886–894.
  • Riesenbeck LM, Bierer S, Hoffmeister I, et al. Hypothyroidism correlates with a better prognosis in metastatic renal cancer patients treated with sorafenib or sunitinib. World J Urol. 2011;29(6):807–813.
  • Schmidinger M, Vogl UM, Bojic M, et al. Hypothyroidism in patients with renal cell carcinoma: blessing or curse? Cancer. 2011;117(3):534–544.
  • Hu W, Wang G, Wang Y, et al. Uncoupling therapeutic efficacy from immune-related adverse events in immune checkpoint blockade. iScience. 2020;23(10): 101580. .
  • Liu X, Shi Y, Zhang D, et al. Risk factors for immune-related adverse events: what have we learned and what lies ahead? Biomark Res. 2021;9(1):79.
  • Maruzzo M, Verzoni E, Vitale MG, et al. Prognostic value of thyroid hormone ratio in patients with advanced metastatic renal cell carcinoma: results from the threefour study (meet-URO 14). Front Oncol. 2021 Nov 25;11:787835. DOI:10.3389/fonc.2021.787835.
  • Pasqualetti G, Schirripa M, Dochy E, et al. Thyroid hormones ratio is a major prognostic marker in advanced metastatic colorectal cancer: results from the phase III randomised CORRECT trial. Eur J Cancer. 2020;133:66–73.
  • Dworakowska D, Grossman AB. Thyroid disease in the time of COVID-19. Endocrine. 2020;68(3):471–474.
  • Sanyaolu A, Okorie C, and Marinkovic A, et al. Comorbidity and its impact on patients with COVID-19 [published online ahead of print, 2020 Jun 25]. SN Compr Clin Med. 2020;2(8):1069-1076.
  • Hariyanto TI, Kurniawan A. Thyroid disease is associated with severe coronavirus disease 2019 (COVID-19) infection. Diabetes Metab Syndr. 2020;14(5):1429–1430.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.