Next Article in Journal
Immune Therapies for Hematologic Malignancies
Next Article in Special Issue
Graves’ Disease during Immune Checkpoint Inhibitor Therapy (A Case Series and Literature Review)
Previous Article in Journal
Genetic Screening for Potential New Targets in Chronic Myeloid Leukemia Based on Drosophila Transgenic for Human BCR-ABL1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Endocrine Toxicities of Antineoplastic Therapy

by
Giulia Puliani
1,2 and
Marialuisa Appetecchia
1,*
1
Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
2
Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(2), 294; https://doi.org/10.3390/cancers13020294
Submission received: 29 December 2020 / Accepted: 12 January 2021 / Published: 14 January 2021
(This article belongs to the Special Issue Endocrine Toxicities of Antineoplastic Therapy)
In recent years, the prognosis of many solid tumors has improved markedly thanks to new treatment strategies, including tyrosine kinase inhibitors (TKIs) and immunotherapy [1]. Despite their clinical relevance, these drugs are burdened with side effects, including endocrine ones, which can become serious and life-threatening if not promptly diagnosed and correctly managed [2]. TKIs act by inhibiting the catalytic activity of many kinases, consequently blocking multiple pathways linked to cell proliferation and survival, angiogenesis, and tumor invasion [3]. Thyroid dysfunctions during TKI therapies are very common, with an estimated incidence of 33.2% [4]. In patients with previous hypothyroidism, initiation of TKI therapy may require an increase in the levothyroxine dose. In patients without prior thyroid dysfunction, the most common side effect is the development of hypothyroidism possibly preceded by thyrotoxicosis due to destructive thyroiditis [5]. The pathogenesis is multifactorial and includes both direct thyroid damage, through the reduction in vascularity, inhibition of thyroid peroxidase activity, and onset of destructive thyroiditis, and peripheral effects, such as increased metabolism and clearance of thyroid hormones [5,6]. Thyroid toxicity has been proposed as a predictor of tumor response to TKIs, predicting longer progression-free survival [4,7].
The endocrine toxicity of TKIs goes beyond thyroid disorders and can also be characterized by metabolic complications, altered electrolyte and bone metabolism, and gonadal impairment. Metabolic complications are frequent when using TKIs, as dyslipidemia is reported in approximately 50% of treated patients and diabetes mellitus is reported in 15–40% of cases [8,9]. Worsening or onset of dyslipidemia has been associated with nearly all TKIs, with the exception of imatinib [10]. TKIs can reduce insulin sensitivity, inducing hyperglycemia [11]. Nilotinib appears to be the TKI with the greatest diabetogenic effect. However, it should be considered that TKIs can also cause hypoglycemia and improvement of previous diabetes mellitus [8].
Similar to TKIs, mammalian target of rapamycin (mTOR) inhibitors are also frequently associated with metabolic complications, such as dyslipidemia (both hypertriglyceridemia and hypercholesterolemia) and diabetes mellitus [12].
Both TKIs and mTOR inhibitors have been associated with electrolyte imbalances. The most common alteration is hyponatremia, due to the syndrome of inappropriate antidiuretic hormone secretion (SIADH) [13]. However, other imbalances, such as magnesium and potassium losses, can also occur if EGFR (epidermal growth factor receptor) is blocked [13].
TKIs can also have an impact on bone metabolism; in fact, many TKIs have been associated with secondary hyperparathyroidism and decreased bone turnover [14]. These alterations in bone turnover could be explained by the inhibition of kinases, such as c-KIT and platelet-derived growth factor receptor A, in osteoclasts and osteoblasts [15].
Immunotherapy aims to stimulate the immune response against neoantigens on the cell surface of cancer cells [16], as tumor cells have been shown to escape immune response by exploiting the inhibitory physiological pathways of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) or cytotoxic T-lymphocyte antigen 4 (CTLA-4) [17]. These drugs have been approved for many types of cancer, but the landscape is constantly expanding with new targets and new indications [1,18]. Taking into account the ability of this class of antineoplastic drugs to enhance the immune response, it is easy to understand the high risk of immune-related adverse events associated with these treatments [19]. In particular, the dysregulation of the immune system and autoimmunity are responsible for many endocrine toxicities.
Thyroid dysfunctions are the most common side effects of immunotherapy, affecting 30% of treated patients. Hypothyroidism is more common in patients treated with PD-1 inhibitors and in combination therapies [20], and although adverse events are usually immune-mediated, it is caused by autoimmune thyroiditis in only 45% of cases [21]. PD-1 inhibitors are also associated with an increased risk of hyperthyroidism, which is usually transient and followed by hypothyroidism, although cases of Graves’ disease and thyroid eye disease have also been described [21].
Contrary to thyroid changes, hypophysitis is more associated with CTLA-4 inhibitors, but again, combination therapy increases the risk [22]. Patients may have both a bulking effect, due to increased pituitary tissue, and hormonal insufficiency: isolated adrenocorticotropic hormone (ACTH) deficiency is more common in patients treated with PD-1 and PD-L1, while CTLA-4 inhibitors usually cause multiple anterior pituitary insufficiency [21]. The concomitant development of diabetes insipidus and anterior pituitary deficiency due to CTLA-4 inhibitors has been rarely described. The development of secondary hypocortisolism could be life-threatening and requires immediate diagnosis and replacement therapy [23]. More rarely, immunotherapy has been associated with primary adrenal insufficiency (0.7% of patients) [20]. While rare, it is important to consider this potential side effect, as it could present with an adrenal crisis [21]. Finally, new onset or worsening of hyperglycemia has been described in up to 2% of patients, rarely associated with diabetic ketoacidosis requiring immediate treatment [21].

1. Old Therapies, New Concerns

Although the interest in endocrine toxicities of antineoplastic therapy has grown after the diffusion of target therapies and immunotherapy, “classical” chemotherapy is not free of endocrine side effects.
The most common complication in premenopausal women is menstrual cycle interruption due to many chemotherapy regimens [24]. Moreover, the toxic effects of chemotherapy on fertility are not always reversible, and the cryopreservation of oocytes for women and of spermatozoa for men should nowadays be part of the treatment strategies of these patients [25]. Sexual dysfunction may be a long-term complication of many cancer treatments, not only in case of direct impact on the pelvic region as for gynecologic cancer or prostate cancer, but also because of the systemic effects of chemotherapy [26,27]. Moreover, in patients affected by breast or prostate cancer, hormonal therapy induces bone loss altering physiological bone turnover and exposing patients to high risk of osteoporosis and fragility fractures [28]. Chemotherapy may also cause electrolyte alterations: platinum compounds can induce sodium, potassium, and magnesium alterations, and alkylating agents and vinca alkaloids are usually associated with SIADH [13].

2. Open Issues

Despite the number of studies on the endocrine toxicity of antineoplastic therapy, many areas of uncertainty remain. First, the pathogenesis of endocrine side effects, especially for TKIs, is not always clear [8,10], and the possible association of different treatments could complicate the clinical presentation and management [29]. Second, endocrine toxicities vary between patients, suggesting that other factors such as pre-existing diseases or genetic aspects can play a role [19], and the comprehension of these confounding factors is necessary. Third, some authors have stated that the Common Terminology Criteria for Adverse Events scale is not very reliable for endocrine toxicities, since the differentiation between grades 2 and 3 is difficult, although necessary, since the clinical approaches are very different [8,30]. Finally, the increase in life expectancy of patients affected by cancer could highlight long-term complications of these treatments, as partially described for cancer survivors.

3. Future Directions: Towards an Oncological Endocrinologist

Considering what has been reported so far, endocrine complications are a clinically significant problem for patients suffering from cancer for two main reasons: the clinical impact of endocrine toxicities, which can be life-threatening and can strongly impair quality of life, and the possible antineoplastic treatment discontinuation, frequent in the case of incorrect or late management of endocrine toxicities.
It is therefore necessary, especially in specialized oncological centers, to include the figure of the endocrinologist with a precise expertise in endocrine toxicities of antineoplastic therapy and to establish a strong collaboration between endocrinologists and oncologists in order to increase awareness on these issues and optimize oncological treatment.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TKItyrosine kinase inhibitor
mTORmammalian targets of rapamycin
TSHthyrotropin-stimulating hormone
SIADHsyndrome of inappropriate antidiuretic hormone secretion
PD-1programmed cell death protein 1
PD-L1programmed cell death protein ligand 1
CTLA-4cytotoxic T-lymphocyte antigen 4
ACTHadrenocorticotropic hormone

References

  1. Tang, J.; Shalabi, A.; Hubbard-Lucey, V.M. Comprehensive analysis of the clinical immuno-oncology landscape. Ann. Oncol. 2018, 29, 84–91. [Google Scholar] [CrossRef] [PubMed]
  2. Ruggeri, R.M.; Campennì, A.; Giuffrida, G.; Trimboli, P.; Giovanella, L.; Trimarchi, F.; Cannavò, S. Endocrine and metabolic adverse effects of immune checkpoint inhibitors: An overview (what endocrinologists should know). J. Endocrinol. Investig. 2018, 42, 745–756. [Google Scholar] [CrossRef] [PubMed]
  3. Krause, D.S.; Van Etten, R.A. Tyrosine Kinases as Targets for Cancer Therapy. N. Engl. J. Med. 2005, 353, 172–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gabora, K.; Piciu, A.; Bădulescu, I.C.; Larg, M.I.; Stoian, I.-A.; Piciu, D. Current evidence on thyroid related adverse events in patients treated with protein tyrosine kinase inhibitors. Drug Metab. Rev. 2019, 51, 562–569. [Google Scholar] [CrossRef] [PubMed]
  5. Lodish, M.B. Clinical review: Kinase inhibitors: Adverse effects related to the endocrine system. J. Clin. Endocrinol. Metab. 2013, 98, 1333–1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Jannin, A.; Penel, N.; Ladsous, M.; Vantyghem, M.C.; Cao, C.D. Tyrosine kinase inhibitors and immune checkpoint inhibitors-induced thyroid disorders. Crit. Rev. Oncol. Hematol. 2019, 141, 23–35. [Google Scholar] [CrossRef] [PubMed]
  7. Rizza, L.; Sbardella, E.; Gianfrilli, D.; Lauretta, R.; Tenuta, M.; Del Bene, G.; Longo, F.; Faggiano, A.; Lenzi, A.; Giannetta, E.; et al. Thyroid profile during the alternative Sunitinib dosing 2/1 schedule in metastatic renal cell carcinoma. Endocrine 2020, 67, 597–604. [Google Scholar] [CrossRef]
  8. Castinetti, F.; Albarel, F.; Archambeaud, F.; Bertherat, J.; Bouillet, B.; Buffier, P.; Briet, C.; Cariou, B.; Caron, P.; Chabre, O.; et al. Endocrine side-effects of new anticancer therapies: Overall monitoring and conclusions. Ann. d’Endocrinol. 2018, 79, 591–595. [Google Scholar] [CrossRef]
  9. Fallahi, P.; Ferrari, S.M.; Elia, G.; Ragusa, F.; Paparo, S.R.; Camastra, S.; Mazzi, V.; Miccoli, M.; Benvenga, S.; Antonelli, A. Therapy of Endocrine Disease: Endocrine-metabolic effects of treatment with multikinase inhibitors. Eur. J. Endocrinol. 2020, 184, R29–R40. [Google Scholar] [CrossRef]
  10. Buffier, P.; Bouillet, B.; Smati, S.; Archambeaud, F.; Cariou, B.; Verges, B. Expert opinion on the metabolic complications of new anticancer therapies: Tyrosine kinase inhibitors. Ann. d’Endocrinol. 2018, 79, 574–582. [Google Scholar] [CrossRef]
  11. Gallo, M.; Muscogiuri, G.; Felicetti, F.; Faggiano, A.; Trimarchi, F.; Arvat, E.; Vigneri, R.; Colao, A. Adverse glycaemic effects of cancer therapy: Indications for a rational approach to cancer patients with diabetes. Metabolism 2018, 78, 141–154. [Google Scholar] [CrossRef]
  12. Vergès, B.; Walter, T.; Cariou, B. ENDOCRINE SIDE EFFECTS OF ANTI-CANCER DRUGS: Effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur. J. Endocrinol. 2014, 170, R43–R55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Verzicco, I.; Regolisti, G.; Quaini, F.; Bocchi, P.; Brusasco, I.; Ferrari, M.; Passeri, G.; Cannone, V.; Coghi, P.; Fiaccadori, E.; et al. Electrolyte Disorders Induced by Antineoplastic Drugs. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef]
  14. O’Sullivan, S.; Lin, J.-M.; Watson, M.; Callon, K.E.; Tong, P.C.; Naot, D.; Horne, A.; Aati, O.; Porteous, F.; Gamble, G.; et al. The skeletal effects of the tyrosine kinase inhibitor nilotinib. Bone 2011, 49, 281–289. [Google Scholar] [CrossRef] [PubMed]
  15. Berman, E.; Nicolaides, M.; Maki, R.G.; Fleisher, M.; Chanel, S.; Scheu, K.; Wilson, B.-A.; Heller, G.; Sauter, N.P. Altered Bone and Mineral Metabolism in Patients Receiving Imatinib Mesylate. N. Engl. J. Med. 2006, 354, 2006–2013. [Google Scholar] [CrossRef] [PubMed]
  16. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [Green Version]
  17. Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [Green Version]
  18. Gong, J.; Chehrazi-Raffle, A.; Reddi, S.; Salgia, R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: A comprehensive review of registration trials and future considerations. J. Immunother. Cancer 2018, 6, 8. [Google Scholar] [CrossRef]
  19. Yang, H.; Yao, Z.; Zhou, X.; Zhang, W.; Zhang, X.; Zhang, F. Immune-related adverse events of checkpoint inhibitors: Insights into immunological dysregulation. Clin. Immunol. 2020, 213, 108377. [Google Scholar] [CrossRef]
  20. Barroso-Sousa, R.; Barry, W.T.; Garrido-Castro, A.C.; Hodi, F.S.; Min, L.; Krop, I.E.; Tolaney, S.M. Incidence of Endocrine Dysfunction Following the Use of Different Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 173–182. [Google Scholar] [CrossRef]
  21. Barnabei, A.; Carpano, S.; Chiefari, A.; Bianchini, M.; Lauretta, R.; Mormando, M.; Puliani, G.; Paoletti, G.; Appetecchia, M.; Torino, F. Case Report: Ipilimumab-Induced Panhypophysitis: An Infrequent Occurrence and Literature Review. Front Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
  22. Haanen, J.; Carbonnel, F.; Robert, C.; Kerr, K.; Peters, S.; Larkin, J.; Jordan, K.; Committee, E.G. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv119–iv142. [Google Scholar] [CrossRef]
  23. Faje, A. Immunotherapy and hypophysitis: Clinical presentation, treatment, and biologic insights. Pituitary 2016, 19, 82–92. [Google Scholar] [CrossRef] [PubMed]
  24. Torino, F.; Barnabei, A.; De Vecchis, L.; Appetecchia, M.; Strigari, L.; Corsello, S.M. Recognizing menopause in women with amenorrhea induced by cytotoxic chemotherapy for endocrine-responsive early breast cancer. Endocr. Relat. Cancer 2012, 19, R21–R33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Oktay, K.; Harvey, B.E.; Partridge, A.H.; Quinn, G.P.; Reinecke, J.; Taylor, H.S.; Wallace, W.H.; Wang, E.T.; Loren, A.W. Fertility Preservation in Patients With Cancer: ASCO Clinical Practice Guideline Update. J. Clin. Oncol. 2018, 36, 1994–2001. [Google Scholar] [CrossRef] [PubMed]
  26. Candy, B.; Jones, L.; Vickerstaff, V.; Tookman, A.; King, M. Interventions for sexual dysfunction following treatments for cancer in women. Cochrane Database Syst. Rev. 2016, 2, CD005540. [Google Scholar] [CrossRef] [PubMed]
  27. Schover, L. Sexual quality of life in men and women after cancer. Climacteric 2018, 22, 553–557. [Google Scholar] [CrossRef] [PubMed]
  28. Handforth, C.; D’Oronzo, S.; Coleman, R.E.; Brown, J.E. Cancer Treatment and Bone Health. Calcif. Tissue Int. 2018, 102, 251–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
  30. Illouz, F.; Briet, C.; Cloix, L.; Le Corre, Y.; Baize, N.; Urban, T.; Martin, L.; Rodien, P. Endocrine toxicity of immune checkpoint inhibitors: Essential crosstalk between endocrinologists and oncologists. Cancer Med. 2017, 6, 1923–1929. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Puliani, G.; Appetecchia, M. Endocrine Toxicities of Antineoplastic Therapy. Cancers 2021, 13, 294. https://doi.org/10.3390/cancers13020294

AMA Style

Puliani G, Appetecchia M. Endocrine Toxicities of Antineoplastic Therapy. Cancers. 2021; 13(2):294. https://doi.org/10.3390/cancers13020294

Chicago/Turabian Style

Puliani, Giulia, and Marialuisa Appetecchia. 2021. "Endocrine Toxicities of Antineoplastic Therapy" Cancers 13, no. 2: 294. https://doi.org/10.3390/cancers13020294

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop