Elsevier

Seminars in Cancer Biology

Volume 79, February 2022, Pages 180-196
Seminars in Cancer Biology

Molecular targets of tyrosine kinase inhibitors in thyroid cancer

https://doi.org/10.1016/j.semcancer.2020.11.013Get rights and content

Abstract

Thyroid cancer (TC) is the eighth most frequently diagnosed cancer worldwide with a rising incidence in the past 20 years. Surgery is the primary strategy of therapy for patients with medullary TC (MTC) and differentiated TC (DTC). In DTC patients, radioactive iodine (RAI) is administered after thyroidectomy. Neck ultrasound, basal and thyroid-stimulating hormone-stimulated thyroglobulin are generally performed every three to six months for the first year, with subsequent intervals depending on initial risk assessment, for the detection of possible persistent/recurrent disease during the follow up. Distant metastases are present at the diagnosis in ∼5 % of DTC patients; up to 15 % of patients have recurrences during the follow up, with a survival reduction (70 %–50 %) at 10-year. During tumor progression, the iodide uptake capability of DTC cancer cells can be lost, making them refractory to RAI, with a negative impact on the prognosis.

Significant advances have been done recently in our understanding of the molecular pathways implicated in the progression of TCs. Several drugs have been developed, which inhibit signaling kinases or oncogenic kinases (BRAFV600E, RET/PTC), such as those associated with Platelet-Derived Growth Factor Receptor and Vascular Endothelial Growth Factor Receptor.

Tyrosine kinase receptors are involved in cancer cell proliferation, angiogenesis, and lymphangiogenesis. Several tyrosine kinase inhibitors (TKIs) are emerging as new treatments for DTC, MTC and anaplastic TC (ATC), and can induce a clinical response and stabilize the disease. Lenvatinib and sorafenib reached the approval for RAI-refractory DTC, whereas cabozantinib and vandetanib for MTC. These TKIs extend median progression-free survival, but do not increase the overall survival. Severe side effects and drug resistance can develop in TC patients treated with TKIs. Additional studies are needed to identify a potential effective targeted therapy for aggressive TCs, according to their molecular characterization.

Introduction

Among endocrine tumors, thyroid cancer (TC) is the eighth most frequently diagnosed cancer worldwide [1], with a rising incidence in the past 20 years [[2], [3], [4], [5], [6], [7]]. Thyroid follicular epithelial-derived cancers, also known as differentiated thyroid carcinomas (DTC), represent over 90 % of TC and are divided into papillary TC (PTC; about 90 %), follicular TC (FTC; about 10 %) and anaplastic TC (ATC, less than 2 %). Medullary TC (MTC) represents less than 5 % of TCs. MTC can be hereditary (25 %), including familial MTC and multiple endocrine neoplasia type 2 (MEN) 2A, MEN 2B, or sporadic (75 %) [8].

The primary therapeutic strategy for patients with DTC and MTC is surgery [9]. In patients with DTC, radioactive iodine (RAI) is administered after thyroidectomy to ablate residual normal thyroid tissue, can be an adjuvant treatment of subclinical micrometastatic disease, and/or treat evident residual or metastatic TC [10]. For the detection of possible persistent/recurrent disease in the follow up, neck ultrasound and basal and thyroid-stimulating hormone (TSH)-stimulated thyroglobulin (Tg) are generally performed every three to six months for the first year, with subsequent intervals depending on initial risk assessment [[11], [12], [13]].

TCs have different clinical behaviors, from indolent tumors with low mortality in most of the cases, to very aggressive forms, i.e. ATC [14]. Over 90 % of DTC patients has a normal life expectancy [15]. At the diagnosis, distant metastases (lung, bone, or other sites) are reported in ∼5 % of DTC patients. In the follow up, about 15 % of patients develop recurrences (in lymph nodes or in thyroid bed), with a survival decrease at 10-years from 70 % to 50 % [16,17]. When DTC progesses, the iodide uptake by thyroid cells is abolished. Consequently, the cells become refractory to RAI, and this impacts negatively the prognosis [[15], [16], [17]]. For patients with metastatic DTC that progresses regardless of systemic treatment, such as RAI and TSH-suppressive thyroid hormone treatment [18], therapeutic options [e.g., surgery, chemotherapy and external beam radiation therapy (EBRT)], are associated with significant side effects and a palliative role (10–20 % transient efficacy), with no prolongation of survival either used alone or in combination [19]. It may be difficult to differentiate ATC from poorly differentiated thyroid cancer (PDTC), which represents an aggressive subtype of TC, but less than ATC [20].

Significant advances have been done recently in the knowledge of the molecular pathways implicated in TC progression. Novel classes of drugs that inhibit oncogenic kinases [e.g., v-Raf murine sarcoma viral oncogene homolog B (BRAFV600E), REarranged during Transfection (RET)/PTC], or signaling kinases [e.g., Vascular Endothelial Growth Factor Receptor (VEGFR), Platelet-Derived Growth Factor Receptor (PDGFR), etc.] associated with TC cell proliferation have been described [[21], [22], [23], [24]]. Most of these molecular alterations are prognostic and diagnostic markers and therapeutical targets [[21], [22], [23]].

During the last years, several small molecule inhibitors that target the intracellular tyrosine kinase (TK) associated with various receptors [i.e., VEGFR2/3, PDGFR, RET, KIT, epidermal growth factor receptor (EGFR), c-Met, fibroblast growth factor receptors (FGFR), and Tie2] have been approved by US Food and Drug Administration (FDA) and European Medicines Agency (EMA) for treating aggressive and refractory TCs [21,22,24]. As expected considering agents that target the ATP-binding site of the intracellular TK, different small molecule TK inhibitors (TKIs) affect multiple TKs beside their initially intended target [25].

In this paper, we review the molecular targets of TKIs, and the results that have emerged from clinical studies that aimed to evaluate the safety and efficacy of these drugs in aggressive TCs.

Section snippets

Gene fusions

The types of gene fusions correlate with the histological subtypes of TC. Thyroid adenoma associated gene fusions (THADA) and paired box 8 (PAX8)/Peroxisome Proliferator-Activated Receptor (PPAR)γ rearrangements are present in follicular variant of PTC (FVPTC), FTC, and follicular adenomas, whereas fusions of TK activating the mitogen-activated protein kinase (MAPK) pathway are reported in PTC [26]. Oncogenic fusions have been shown in 6–46 % of sporadic PTC [[27], [28], [29]]. RET, PPARG,

In vivo studies with TKIs in TC (Table 1)

An emerging potentially effective option for the treatment of advanced TCs is represented by TKIs. TKIs are multikinase inhibitors (mTKI) affecting multiple TKs in addition to their initial intended targets. mTKI interfere with some of the abovereported pathways, involved in several mechanisms that favor tumor growth and dissemination, (e.g., angiogenesis, invasiveness, and local and distant spread) [119]. Fig. 1 schematically illustrates the human receptor tyrosine kinases involved in TCs. The

Conclusions and perspectives

TC is the eighth most frequently diagnosed cancer worldwide [1] and its incidence has raised in the past 20 years. Surgery is the primary strategy of treatment for patients with DTC and MTC [9]. Neck ultrasound, basal and TSH-stimulated Tg are generally performed every three to six months for the first year, for the detection of possible persistent/recurrent disease during the follow up [[11], [12], [13]]. For patients with metastatic DTC that progresses despite systemic treatment, such as RAI

References (215)

  • G. Varricchi et al.

    Innate effector cells in angiogenesis and lymphangiogenesis

    Curr. Opin. Immunol.

    (2018)
  • G. Varricchi et al.

    Angiogenesis and lymphangiogenesis in inflammatory skin disorders

    J. Am. Acad. Dermatol.

    (2015)
  • G. Bunone et al.

    Expression of angiogenesis stimulators and inhibitors in human thyroid tumors and correlation with clinical pathological features

    Am. J. Pathol.

    (1999)
  • C.M. Lennard et al.

    Intensity of vascular endothelial growth factor expression is associated with increased risk of recurrence and decreased disease-free survival in papillary thyroid cancer

    Surgery

    (2001)
  • A. Antonelli et al.

    Novel therapeutic clues in thyroid carcinomas: the role of targeting cancer stem cells

    Med. Res. Rev.

    (2017)
  • M. Blomberg et al.

    Thyroid cancer in Denmark 1943-2008, before and after iodine supplementation

    Int. J. Cancer

    (2012)
  • J. Albores-Saavedra et al.

    Changing patterns in the incidence and survival of thyroid cancer with follicular phenotype – papillary, follicular, and anaplastic: a morphological and epidemiological study

    Endocr. Pathol.

    (2007)
  • Y. Wang et al.

    Increasing incidence of thyroid cancer in Shanghai, China, 1983-2007

    Asia. J. Public Health

    (2015)
  • T. Carling et al.

    Thyroid cancer

    Annu. Rev. Med.

    (2014)
  • J.S. Lin et al.

    Screening for thyroid cancer: updated evidence report and systematic review for the US preventive services task force

    JAMA

    (2017)
  • M.P. Pusztaszeri et al.

    Update on the cytologic and molecular features of medullary thyroid carcinoma

    Adv. Anat. Pathol.

    (2014)
  • P. Miccoli et al.

    Completion total thyroidectomy in children with thyroid cancer secondary to the Chernobyl accident

    Arch Surg

    (1998)
  • L. Lamartina et al.

    Radioiodine remnant ablation in low-risk differentiated thyroid cancer: the "con" point of view

    Endocrine

    (2015)
  • A. Antonelli et al.

    Role of neck ultrasonography in the follow-up of patients operated on for thyroid cancer

    Thyroid

    (1995)
  • A. Antonelli et al.

    Role of neck ultrasonography in the follow-up of children operated on for thyroid papillary cancer

    Thyroid

    (2003)
  • American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer et al.

    Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer

    Thyroid

    (2009)
  • C. Durante et al.

    Papillary thyroid cancer: time course of recurrences during postsurgery surveillance

    J. Clin. Endocrinol. Metab.

    (2013)
  • J.J. Gruber et al.

    Differentiated thyroid cancer: focus on emerging treatments for radioactive iodine-refractory patients

    Oncologist

    (2015)
  • M. D’Agostino et al.

    Different expression of TSH receptor and NIS genes in thyroid cancer: role of epigenetics

    J. Mol. Endocrinol.

    (2014)
  • P. Miccoli et al.

    Prospective, randomized, double-blind study about effectiveness of levothyroxine suppressive therapy in prevention of recurrence after operation: result at the third year of follow-up

    Surgery

    (1993)
  • K.D. Burman

    Is poorly differentiated thyroid cancer poorly characterized?

    J. Clin. Endocrinol. Metab.

    (2014)
  • S.M. Ferrari et al.

    Lenvatinib in the therapy of aggressive thyroid cancer: state of the art and new perspectives with patents recently applied

    Recent Pat. Anticancer Drug Discov.

    (2018)
  • S.M. Ferrari et al.

    Sunitinib in the treatment of thyroid cancer

    Curr. Med. Chem.

    (2019)
  • Y.E. Nikiforov et al.

    Molecular genetics and diagnosis of thyroid cancer

    Nat. Rev. Endocrinol.

    (2011)
  • A. Antonelli et al.

    New targeted therapies for thyroid cancer

    Curr. Genomics

    (2011)
  • V.D. Yakushina et al.

    Gene fusions in thyroid cancer

    Thyroid

    (2018)
  • M.L. Prasad et al.

    NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States

    Cancer

    (2016)
  • Z. Lu et al.

    Targeted next generation sequencing identifies somatic mutations and gene fusions in papillary thyroid carcinoma

    Oncotarget

    (2017)
  • A. Zehir et al.

    Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients

    Nat. Med.

    (2017)
  • Y.E. Nikiforov

    Thyroid carcinoma: molecular pathways and therapeutic targets

    Mod. Pathol.

    (2008)
  • V. Marotta et al.

    RET/PTC rearrangement in benign and malignant thyroid diseases: a clinical standpoint

    Eur. J. Endocrinol.

    (2011)
  • J.A. Fagin

    Challenging dogma in thyroid cancer molecular genetics–role of RET/PTC and BRAF in tumor initiation

    J. Clin. Endocrinol. Metab.

    (2004)
  • M. Santoro et al.

    RET/PTC activation in papillary thyroid carcinoma: european journal of endocrinology prize lecture

    Eur. J. Endocrinol.

    (2006)
  • A. Antonelli et al.

    RET TKI: potential role in thyroid cancers

    Curr. Oncol. Rep.

    (2012)
  • M. Xing

    BRAF mutation in thyroid cancer

    Endocr. Relat. Cancer

    (2005)
  • H.M. Rabes et al.

    Pattern of radiation-induced RET and NTRK1 rearrangements in 191 post-chernobyl papillary thyroid carcinomas: biological, phenotypic, and clinical implications

    Clin. Cancer Res.

    (2000)
  • R.J. Leeman-Neill et al.

    RET/PTC and PAX8/PPARγ chromosomal rearrangements in post-chernobyl thyroid cancer and their association with iodine-131 radiation dose and other characteristics

    Cancer

    (2013)
  • A. Antonelli et al.

    Epidemiologic and clinical evaluation of thyroid cancer in children from the Gomel region (Belarus)

    World J. Surg.

    (1996)
  • X. Su et al.

    Radiation exposure, young age, and female gender are associated with high prevalence of RET/PTC1 and RET/PTC3 in papillary thyroid cancer: a meta-analysis

    Oncotarget

    (2016)
  • M. Erdoğan et al.

    The prevalence of RET/PTC mutations in papillary thyroid cancers in Turkish population and its relation between tumor histopathology and prognostic factors

    Exp. Clin. Endocrinol. Diabetes

    (2008)
  • Cited by (71)

    • Molecular basis and targeted therapy in thyroid cancer: Progress and opportunities

      2023, Biochimica et Biophysica Acta - Reviews on Cancer
    View all citing articles on Scopus
    View full text