Open access peer-reviewed chapter

RET Proto-Oncogene Mutations: Impact on Diagnosis, Treatment and Prognosis of MTC

Written By

Shabnam Heydarzadeh and Mehdi Hedayati

Submitted: 16 July 2022 Reviewed: 08 November 2022 Published: 17 April 2023

DOI: 10.5772/intechopen.108941

From the Edited Volume

Thyroid Cancer - The Road From Genes to Successful Treatment

Edited by Ifigenia Kostoglou-Athanassiou

Chapter metrics overview

79 Chapter Downloads

View Full Metrics

Abstract

Variants of MTC result from different mutations in exons of the RET gene. RET proto-oncogene is activated by a DNA rearrangement and it is one of the first tyrosine kinase receptor (RTK) proteins found to play a role in neoplasia. Early detection using genetic screening has become the gold standard of therapy, followed by prophylactic thyroidectomy. RET-kinase inhibitors have been developed recently for the treatment of MTC and are currently at various phases of pre- and clinical trials. Numerous autosomal dominantly inherited mutations have been demonstrated to activate RET constitutively. These mutations in separate populations are believed to be correlated with a rather heterogeneous prototype across countries. As such, one objective of this study was to demonstrate a geographical pattern of RET mutations in various populations. Advances in RET genetic screening have facilitated for the rapid recognition of hereditary MTCs and prophylactic thyroidectomy for relatives who may not show signs of the disease. In this chapter, we will discuss oncogenic RET signaling, RET inhibitors and the major RET mutations found in MTC and the necessity of RET genetic screening for the early diagnosis of MTC patients, using American Thyroid Association guidelines and genotype-phenotype correlation.

Keywords

  • medullary thyroid cancer
  • RET proto-oncogene
  • RET mutation
  • RET signaling
  • RET inhibitors

1. Introduction

The medullary thyroid carcinoma (MTC) is one of the most aggressive kinds of thyroid cancer. It is a neuroendocrine tumor and is notably distinct from differentiated thyroid carcinoma. MTC accounts for 5–10% of all thyroid malignancies and occurs in both sporadic (75%) and inherited (25% of cases) forms [1, 2]. The latter exhibits an autosomal dominant inheritance pattern with varying expressivity and age-dependent penetrance [3, 4]. RET (REarranged during Transfection) proto-oncogene is essential for the molecular pathogenesis of hereditary MTCs [5].

The human RET proto-oncogene encodes a transmembrane receptor tyrosine kinase that transmits growth and differentiation signals. Extracellular binding of ligands and coreceptors, receptor dimerization via the cysteine-rich domain, and intracellular autophosphorylation of the tyrosine kinase catalytic domain are required for RET function. RET can be activated oncogenically in vivo and in vitro by cytogenetic rearrangement. RET gene mutations have been identified in a variety of human disorders, including PTC (Papillary thyroid cancer), MTC, MEN2A, and MEN2B. Consequently, RET is referred to as the one implicated in numerous disorders [6, 7]. RET proto-oncogene is known as a molecular therapeutic target in thyroid cancer. Numerous factors, including as sex, age, tumor stage, and tumor grade can influence the prognosis. Therefore, the average survival rate for patients with thyroid gland cancers (95 percent) is approximately 10 years. Patients with regional and metastatic illness stages are estimated to have an overall survival rate of 75%, according to estimates. When a patient is diagnosed with distant metastasis, the prognosis is poor, with a 10-year survival rate of only 40% [8, 9]. Extra thyroidal metastasis and stage upon diagnosis are the only independent predictors of MTC patients' life expectancy. In other words, those detected at an early stage and patients without detectable recurrence had a life expectancy [10]. Postoperative calcitonin level and tumor extension have also been identified as significant prognostic variables for identifying MTC patients at high risk for disease recurrence [11]. We proposed that RET genetic mutations may be different in distinct populations. Therefore, in our recent study we found a geographical pattern of RET mutations in different populations [12].

This chapter is a summary of the current understanding of RET mutations and the most advanced therapeutic methods for RET-dependent thyroid tumors. We will discuss oncogenic RET signaling, RET inhibitors, and the major RET mutations found in MTC, as well as the necessity of RET genetic screening for the early diagnosis of MTC patients, in accordance with American Thyroid Association guidelines and genotype-phenotype correlation.

1.1 RET protein kinase structure and activation mechanism

RET is predominantly expressed in peripheral enteric, sympathetic, and sensory neurons, in addition to central motor, dopamine, and noradrenaline neurons. It is also expressed in branching ureteric buds and differentiating spermatogenia during embryogenesis [13]. RET contains three distinct transcripts, each of which encodes RET isoforms. RET exon 19 is present in all transcripts; however, the 3' end of exon 19 undergoes variable splicing, resulting in transcripts in which exon 19 is unspliced, spliced to exon 20, or spliced to exon 21. These transcripts encode RET isoforms with 9 (RET9), 51 (RET51), or 43 (RET43) amino acid c-terminal ends. RET9 and RET51, composed of 1072 and 1114 amino acids, are the predominant isoforms in vivo. These two isoforms are co-expressed in the majority of tissues but have differential developmental roles and gene expression profiles, suggesting possible discrepancies in cell-cell contact pathway regulation [14].

Tyrosine (Y1062), the last amino acid shared by all three isoforms, is phosphorylated during RET activation. Thus, alternate splicing inserts Y1062 in distinct contexts of amino acids in the three RET isoforms, imparting distinct binding potentials. An N-terminal extracellular portion of RET contains a ligand-binding domain, a cadherin (Ca2+-dependent cell adhesion)-like domain, and a cysteine-rich domain (near the cell membrane). This domain is a ligand for glial cell-derived neurotropic factor (GDNF), an activator protein [15]. A hydrophobic transmembrane domain and an intracellular TK domain are the other two domains. The TK domain contains several tyrosine residues (16 in RET9 and 18 in RET51), two of which are unique to RET51 at locations 1019 and 1051. The transmembrane domain ensures the close proximity of RET monomers via noncovalent interactions between receptors. Two TK subdomains, which are phosphorylated upon receptor activation and are important in the activation of intracellular signaling pathways, are present in the intracellular region [16, 17].

GDNF, NRTN, ARTN, and PSPN are ligands of the RET receptor TK that belong to GFLs. RET is unphosphorylated and inactive in the absence of these ligands. Multiple signaling pathways are activated as a result of the activation of receptor dimerization and autophosphorylation caused by the binding of ligand to the extracellular domain of the RET receptor by GFR co-receptors [18]. In other words, after GFL binds to the RET receptor, an intracytoplasmic domain within the upstream portion of RET is autophosphorylated, stabilizing the protein and necessitating subsequent downstream activity of the RET autophosphorylation cascade. In fact, phosphorylation of Tyr981, in addition to Tyr1015, Tyr1062, and Tyr1096, is crucial for beginning intracellular signal transduction cascades [19]. It is believed that RET signaling provides growth and survival signals through the RAF-MEK-ERK and PI3K-AKT-mTOR pathways [20, 21].

1.2 Intracellular signaling pathway of RET mutations

The RET gene is located on chromosome 10q11.2, is approximately 55,000 base pairs in length, includes 21 exons, and encodes a single-pass transmembrane receptor tyrosine kinase (RTK) that is mostly expressed in neural crest and urogenital tract precursor cells [10, 22]. The RET proto-oncogene encodes a receptor tyrosine kinase with four cadherin-related motifs and a cysteine-rich region in the extracellular domain, and its four ligands mentioned above. When these neurotrophic factors are administered, they activate a unique receptor system that consists of the GFR1–4 coreceptor, which is the receptor for the ligand-binding component, and the GFR2–4 coreceptor, which is responsible for the signaling component [19, 23].

Alternate 3'-spicing generates three splicing variants of RET, including RET9, RET43, and RET51. Of these, RET9 and RET51 have the most significant isoforms, each with 1072 amino acids. Through the GFR1–4 (GDNF family receptors 1–4), GFL activation of RET can be induced. These ligands activate intrinsic tyrosine kinase activity when they interact with GFR1–4 [20, 23]. In order to activate RET, the ligand must first form a complex with the necessary co-receptor. This co-receptor then interacts on the cell membrane with the RET protein, which leads to the dimerization of the receptor and the beginning of intracellular signaling via the tyrosine kinase domains [24].

Oncogenic RET proteins activate a complex network of signal transduction pathways that contributes to cellular transformation. Binding of the ligand GFR complex to RET triggers its homo dimerization, phosphorylation of tyrosine residues and subsequent intracellular signaling; subsequently, RET activation leads to increased proliferation through a complex network of second messengers, and the molecular partners and/or targets include Jun N-terminal kinase (JNK); mammalian target of rapamycin (m-TOR); phosphatidyl- inositol 3 kinase (PI3K), son of seven less (SOS); vascular endothelial growth factor (VEGF); growth actor receptor bound protein 2 (GRB2), hypoxia inducible factor 1a (HIF1a), extracellular signal-regulated kinase (ERK), protein kinase C (PKC), pyruvate dehydrogenase kinase (PDK), phospholipase Cγ (PLCγ) [24].

The intracellular domain of RET contains autophosphorylation sites, and phosphorylated tyrosine serve as docking sites for signaling molecules [25, 26]. Phosphorylated tyrosine 1062, also known as Y1062, is one of these residues. It serves as a binding site for several different adaptor proteins, including Shc, FRS2, Dok1/4/5, IRS1/2, and Enigma, and it is critical to the capacity of mutant RET to transform cells. In addition, it was discovered that tyrosine 905 binds to Grb7/10, tyrosine 981 binds to Src, tyrosine 1015 binds to phospholipase Cγ (PLCγ), and tyrosine 1096 binds to Grb2; all of these findings were independently confirmed by other researchers [27]. Interestingly, RAS/ERK, (PI3K)/AKT, p38MAPK, and JNK pathways are activated mainly through tyrosine 1062. When the adaptor protein Shc binds to phosphorylated tyrosine 1062, it recruits the Grb2-Gab1 and Grb2-Sos complexes that then activate the PI3K/AKT and RASERK pathways, respectively (Figure 1) [23, 28].

Figure 1.

RET Intracellular signaling pathway. homodimeric GFLs activate the transmembrane RET tyrosine kinase by binding to different GFR receptors; binding of the ligand GFRα complex to RET initiate the homodimerization, phosphorylation of tyrosine residues and subsequent intracellular signaling. RET activation leads to high proliferation through a complex-network of second and third messengers that is developmentally-dependent and tissue-specific.

1.3 Oncogenic RET inhibitors

Preclinical models and early phase clinical trials have explored targeted therapy through inhibition of RET and downstream signaling pathways. Phase III trials of the multi kinase inhibitors Vandetanib and Cabozantinib showed improvement in PFS (progression-free survival) but with many adverse events, which led to a trial of lower- dose Vandetanib [29].

In recent years, selective RET inhibitors have been created in an effort to obtain increased potency while also achieving lower toxicity (Figure 2). Pralsetinib (BLU-667) and Selpercatinib (LOXO-292) are examples of such next-generation small molecule inhibitors that have been rapidly developed and introduced into clinical testing. Both inhibitors are capable of blocking a wide spectrum of RET changes, including M918T, C634W, gatekeeper mutations V804L and V804M, KIF5B-RET, and CCDC6-RET, according to the functional tests that were conducted utilising a variety of in vitro and in vivo models. It is important to note that LOXO-292 and BLU-667 have substantially less activity against VEGFR2 in comparison to modifications in RET, which could potentially reduce their toxicity [30].

Figure 2.

Oncogenic RET signaling and selective RET inhibitors.

The RET receptor can be inhibited effectively and selectively by LOXO-292. Both RET mutations, as observed in MTC, and RET fusions are the targets of this medication (seen in PTC, PDTC, and ATC). The RET V804 gatekeeper mutation, which is linked to resistance to RET-targeted kinase inhibitors, was the primary focus of the research that went into the development of this medication. Because LOXO-292 is able to pass the blood-brain barrier and achieve therapeutic concentrations in the central nervous system, it has the potential to be used as a therapy for brain metastases caused by RET mutation or fusion. At the annual meeting of the American Society of Clinical Oncology in 2018, preliminary findings from the LIBRETTO-001 phase 1 dose escalation and expansion trial were presented. Fatigue ranging from grade 1 to 2, diarrhea, constipation, dry mouth, nausea, and dyspnea were the side effects that occurred the most frequently (10 percent to 20 percent). Asymptomatic increase of the alanine aminotransferase level and a case of tumor lysis syndrome were the two conditions in question here. The maximum dose that the patient could tolerate was not achieved. The LIBRETTO-001 clinical trial is currently in the expansion phase, during which additional patients with RET-mutated MTC and RET-fusion malignancies, such as PTC (papillary thyroid cancer), PDTC (poorly differentiated thyroid cancer), and ATC (anaplastic thyroid cancer), are being enrolled.

Additionally, BLU-292 is an extremely selective and highly effective RET inhibitor. It works in a manner very similar to that of LOXO-292, in that it targets RET fusions as well as RET mutations, such as the RET V804 mutation. A phase 1 escalation/expansion clinical trial of BLU-667 is currently being conducted, and the results of the trial were reported at the annual meeting of the American Association for Cancer Research in 2018. This study’s objectives include determining the maximum tolerated dose, assessing safety, analyzing pharmacokinetics, and evaluating preliminary anticancer activity. Constipation, elevated alanine aminotransferase and aspartate aminotransferase, hypertension, leukopenia, headache, sleeplessness, and exhaustion were the only side effects of the low toxicity that was seen [31].

BOS172738, TPX- 0046, and TAS0953/HM06 are likewise in the early phases of development as selective RET inhibitors. In addition to the RET V804M gatekeeper mutation, multiple alternative pathways of acquired resistance to MKIs have been described. Mechanisms of resistance to selective RET remain a major field of study. According to a preclinical investigation, the unique solvent front mutation KIF5B-RET G810R may develop on-target resistance to Selpercatinib and Pralsetinib, however it remains vulnerable to TPX-0046, a selective RET inhibitor built with a macrocyclic structure to target active RET confirmation [30].

In conclusion, over the past three decades, the involvement of RET activating mutations and rearrangements in carcinogenesis has been proven. With the emergence of extremely selective RET inhibitors, there is significant enthusiasm in the RET sector. In preliminary phase I/II trials, the next-generation selective RET inhibitors Selpercatinib and Pralsetib displayed excellent clinical efficacy and safety. Both agents have obtained breakthrough designations from the FDA. Unanswered questions include the PFS, DOR (duration of response), and OS (overall survival) with these drugs; if all RET aberrant tumors respond similarly for a tissue-agnostic indication; and the mechanisms of acquired resistance to the potent RET inhibitors. In addition, combination therapies that investigate the simultaneous inhibition of RET and associated pathways will shed light on the clinical efficacy of such techniques [30].

According to clinical and preclinical studies, initiation of RET kinase activity has been characterized as a target for a number of tyrosine kinase inhibitors [32]. The finding of molecular targets in thyroid cancer has led to the development of treatments for patients who have advanced forms of the disease. These treatments include FDA-approved drugs such as Cabozantinib and Vandetanib for MTC and Sorafenib and Lenvatinib for differentiated thyroid cancer [33, 34].

Strong inhibition of the target proteins VEGFR-2, MET, RET, KIT, AXL, and TIE2 is provided by Cabozantinib. Because of its potent ability to inhibit RET; Cabozantinib was identified as a particularly promising candidate for treatment in MTC patients. Cabozantinib, in contrast to Vandetanib, does not reduce EGFR activity to a significant degree. Other tyrosine kinase receptor inhibitors, such as ZD6474, which are medicines that are active when taken orally, have an effect on VEGFR-2 and limit the actions of RET tyrosine kinase. In patients with metastatic familial MTC who were participating in a clinical research, it was revealed that ZD6474 therapy triggered some degree of cure [12].

1.4 RET proto-oncogene mutations

There have been a total of 100 different mutations found in the RET gene so far, and with the exception of a few that cause dual phenotypes, the majority of them can be classified as either having a loss of function or a gain of function. Gain-of-function mutations in RET are primarily what cause RET-related malignancies, and these mutations may be broken down into two categories: those that modify cysteine residues in the cysteine-rich domain, and those that alter residues in the RET-KD. Within the first group, the mutated residue that occurs most frequently in MEN2A patients is Cys634. This occurs because the removal of one-half of an intra-molecular disulfide bond makes it possible to form an intermolecular disulfide bond with a second mutant molecule. This results in constitutive receptor dimerization and aberrant signaling. It is not known if activating mutations within RET-KD directly lead to constitutive dimer formation or whether the mechanism for activating mutations is more diverse. RET transformation can be produced by a wide variety of mutations, including L790F, Y791F, S891A, and R844L, but the resulting symptoms are only moderately severe MTC and MEN2A. In contrast, the M918T mutation has a very high capacity for transformation and is present in 95% of MEN2B patients. This mutation is responsible for the disease. A number of mucosal, ophthalmic, and skeletal disorders are included in the MEN2B phenotype. In addition to the thyroid and adrenal glands, this phenotype affects the skeleton. In stark contrast to the MEN2A dimerizing mutations, in which Tyr905 is necessary for oncogenesis, the M918T RET mutation does not require this residue in order to become activated [35]. This implies that various underlying mechanisms disrupt RET activation’s normal control in MEN2A and MEN2B. Furthermore, M918T RET specifically targets novel substrates like STAT3 that may aid in cell transformation [36]. In addition to transforming mutations that occur inside intact RET, chromosome translocations have the potential to produce oncogenic fusions that include the RET kinase domain (RET/PTC oncogenes). These oncogenes are responsible for the development of PTC. RET/PTC fusion proteins are found in the cytoplasm and contain RET-KD from the beginning of exon 12 (which begins at Glu713) all the way through the C terminus. The N-terminal domain of RET/PTC is often a dimerization domain derived from the fusion partner in many instances. Notably, reducing the converting potential of RET/PTC by mutating the residue that corresponds to Tyr905 in wild-type RET results in less transformation [37, 38].

As previously mentioned, RET missense mutations in the germline are linked to MEN2A, MEN2B, and FMTC, whereas sporadic MTC is thought to result from a somatic mutation in the tumor cells, RET mutations are primarily missense and located in exons 10, 11, 13, 14, 15, and 16 (RET’s extracellular domain) (in the TK domain) [5, 39, 40]. A ligand-independent dimerization of receptor molecules, increased phosphorylation of intracellular substrates, and cell transformation can be caused by a mutation of the extracellular cysteine in codon 634 of exon 11 of RET. A mutation in the intracellular TK (for example, codon 918) has no effect on receptor dimerization, but it does promote constitutive activation of intracellular signaling pathways, which in turn culminates in cellular transformation [21, 41].

Exons 10 and 11 have the FMTC-specific mutations as well. Exon 8 (codons 532 and 533), exon 13, (codons 768, 790, and 791), (codons 804 and 844), (codon 891), and exon 16 have also been shown to have non-cysteine point mutations (codon 912) [42]. According to a recent meta-analysis, 39 distinct RET germline mutations have been discovered in FMTC patients from various families since 1993. All mutations were missense type and dispersed among exons 5, 8, 10, 11, 13, 14, 15, and 16 with the exception of a 9-bp duplication (after codon 531, exon 8). In FMTC, age-specific penetrance of cancer growth and nodal metastasis were strongly linked with particular germline RET mutations [43]. Overall, mutations in codons 609, 611, 618, and 620 of exon 10, codon 768 of exon 13, and codon 804 of exon 14 are most frequently related with FMTC. When FMTC is related with mutations in codon 634 of exon 11, C634R is nearly never observed, while C634Y is the most prevalent variant [21].

1.5 Germline screening of RET mutations

The genetic testing for RET germline mutation has demonstrated 100 percent sensitivity and specificity in identifying persons at risk for developing MTC. In comparison to the current standard of annual biochemical monitoring, such as blood calcitonin, this genetic assay allows for earlier and more conclusive diagnosis and clinical management of people who have a familial risk for MTC. Once a person is identified as having a RET mutation, they must receive thorough counseling. In order to give a preventive thyroidectomy to asymptomatic individuals who are diagnosed as RET mutation carriers, it is necessary to identify and test at-risk family members [4].

Since prophylactic thyroidectomy can prevent hMTC, the American Thyroid Association suggests that all patients with MTC be offered germline RET testing [44]. Based on a model that categorizes mutations into risk levels using genotype-phenotype correlations, recommendations for the scheduling of prophylactic thyroidectomy and the extent of surgical resection are made (A-D). The highest risk of MTC is associated with ATA level D (ATA-D) mutations. Codons 883 (exon 15) and 918 (exon 16) are two of these mutations that are linked to the lowest age of onset, the highest risk of metastasis, and the highest fatality rate. A lower but still significant prevalence of aggressive MTC is linked to ATA level C (ATA-C) mutations, which include codon 634 changes (exon 11). ATA-B mutations, which include mutations at codons 609, 611, 618, 620 (exon 10) and 630, are associated with a decreased risk for severe MTC mutations (exon 11). ATA-A mutations are associated with the “least severe” risk. When they have preventative thyroidectomy at age 4 years, these patients have lower serum calcitonin levels, a lower tumor stage, and a better rate of biochemical cure compared to ATA-B mutation carriers of the same age [45]. RET mutations can be found at codons 768, 790, 791 (exon 13), 804 (exon 14), and 891 in ATA-A mutations (exon 15). ATA made the decision to develop specialized MTC Clinical Guidelines in order to compile and update the vast amount of MTC-related literature, as well as to integrate this information with evidence-based medicine and the feedback of a panel of experienced physicians [21, 46].

There are limited reports of these mutations in Iranian families with MTC in the literature [47, 48]. In our recent study, we tested individuals with MTC and their MTC-affected first-degree relatives for RET exon10 mutations. In our latest investigation, 14 individuals with sMTC and FMTC were found to have six distinct mutations in exon10 of RET that were confined to codons 611, 618, and 620, but not codon 609. This data revealed an atypical distribution of RET exon10 mutations in comparison to other groups. In our study population, exon10 of the RET proto-oncogene was mutation-free in MEN2A, MEN2B, and pheochromocytoma. However, exon10 mutations in MEN2A have been found in numerous populations. C611Y and C620R were the most prevalent mutations in exon10 among patients with FMTC and sMTC, respectively [49, 50].

Codon 620 of exon 10 has generally been shown to include eight different variants, including seven missense mutations and one synonymous mutation. The codon in exon 10 with the greatest frequency of mutations during our analysis was codon 620. In other words, more over 50% of the mutations in our investigation were caused by codon 620. Additionally, neither synonymous nor nonsense mutations in exon 10 of the RET proto-oncogene were found in our study population. None of the cysteine codons in exon 10 had any mutations. The findings of this study suggest that mutations in exon 10 of the RET proto-oncogene are limited to three critical cysteine codons (611, 618, and 620), which were only identified in Iranian patients with FMTC and likely sMTC. All patients with exon 10 mutations, with the exception of one, had the haplotype G691S/S904S. In the current analysis, no mutation in the RET proto-exon oncogene’s 10 in the syndromic type of MTC was found [50].

Since the research of other exons within the same gene has received less attention, we investigated the incidence of germ line mutations in exon 2 of the RET proto-oncogene in Iranian patients with MTC. The RET gene has the nucleotide substitutions c135G>A/A45A (rs1800858) in exon 2 and c.337+9G>A (rs2435351) and c.337+137G>T (rs2505530) in the intronic region. Among patients and relatives, the genotype and allele frequencies with the highest and lowest frequencies, respectively, were c.337+137G>T (rs2505530) and c135G>A/A45A (rs1800858). Also, no link was found between identified nucleotide alterations and disease phenotype, gender, or race. No mutations resulting in altered amino acid sequences in exon 2 or exon-intron splice sites were identified. However, additional research is advised to determine the likely correlation between discovered variants and the presence or absences of other mutations in other RET major exons, as well as to determine the haplotype association with the disease [51].

217 people were included in order to study the spectrum of prominent RET germline mutations in exons 10, 11, and 16 in hereditary MTC in the Iranian population. Leukocytes' genomic DNAs were isolated utilizing the Salting Out/Proteinase K technique. The mutations were detected using PCR-RFLP and DNA sequencing. In 217 subjects, 43 missense mutations were found in exons 10, 11, and 16 (6 percent, 13 percent, and 16 percent, respectively) (0.9 percent). In addition, a new germline mutation was found in exon 11 (S686N). In addition, eight individuals had four distinct SNPs in intron 16. The data revealed the frequency profile of RET mutations in Iranian patients with MTC (19.8 percent). In our population, C634G was the most prevalent mutation, but in most populations it was C634R. Collectively, these data highlight the significance of the genetic background of family members of any MTC patient [5].

Finally, it is advised that other RET exons, particularly those with a high frequency of mutations, such as exons 13, 14, and 15, be studied. Additionally, direct sequencing analysis is a reliable tool for detecting unknown RETS mutations. In addition, the transformative activity and functional effect(s) of novel RET mutations such as S686N and intronic polymorphisms have yet to be determined (Figure 3) [5].

Figure 3.

The allele frequency of the RET proto-oncogene mutations in Iranian patients with MTC.

1.6 The relationship between RET tyrosine kinase inhibition and MTC treatment

Recent RET-kinase inhibitors for the treatment of MTC are through various levels of preclinical and clinical testing [52]. A group has launched a phase II clinical research assessing the efficacy of oral ZD6474 (Zactima®) in patients with locally advanced or metastatic MTC: of the 20 patients accrued to date, around 30% have seen objective remissions. Other inhibitors of RET activity targeting various areas of its molecular biology and signaling pathway are in development [24, 53, 54]. The most important drugs for MTC treatment is listed in Figure 4.

Figure 4.

The most important FDA-approved drugs for MTC according to their approved years or current phases of clinical trials.

Patients with MTC are evaluated using tumor markers (calcitonin and carcinoembryonic antigen; CEA), a complete and precise ultrasonography of the neck, and genetic testing. Cross-sectional imaging may be obtained for surgical planning or when suspected distant metastases are present. Biochemical testing is required to exclude primary hyperparathyroidism and PHEO (pheochromocytoma). After this, a total thyroidectomy with central neck dissection is often advised, and in rare instances, more extensive surgery may be required (if indicated by the preoperative assessment). External beam radiation therapy may improve loco regional control if the patient has a high risk of recurrence [34]. Since the response to first therapy influences survival and recurrence risk, an experienced multidisciplinary team should be included from the start [55]. Many of these concerns are addressed in the new American Thyroid Association (ATA) guidelines for MTC management [56].

Watchful waiting, surgery, radiation, cryo ablation, and chemoembolization are treatment options for asymptomatic residual, recurrent, and distant metastatic disease in MTC and differentiated thyroid carcinoma (DTC). In order to minimize skeletal-related occurrences in MTC patients, receptor activator of nuclear factor kappa B ligand (RANKL) inhibitors or intravenous bisphosphonate are often administered [12, 57].

Advertisement

2. ATA Recommendations for MTC management

Since hMTC can be prevented by prophylactic thyroidectomy, the American Thyroid Association (ATA) suggests that all patients with MTC should be offered germline RET mutation testing for mutation discovery and improved patient and family treatment (particularly in RET-positive cases) [44, 56]. Nevertheless, the updated ATA has altered the existing risk categories for hMTC. It is now recommended to undertake genotype to phenotype correlation of disease in order to identify mutations that enhance the risk levels of patients with MTC, hence determining the necessity of prophylactic thyroidectomy and the extent of surgical resection.

Recommendations for the scheduling of prophylactic thyroidectomy and surgical area resection are derived on genotype-phenotype correlations used to categorize mutation risk levels. The preceding ATA advice categorized risk based on four mutation levels: A, B, C, and D. ATA-D mutations, especially codons 883 (exon 15) and 918 (exon 16), were related with the lowest age of onset, the greatest incidence of metastasis, and the highest fatality rate. In the most recent amended ATA guideline, a new “highest risk” (HST) category has been created, which covers patients with MEN2B and the RET codon M918T mutation [46].

ATA-C mutations, including mutations at codon 634 (exon 11), were assumed to provide a decreased risk of aggressive MTC. This category has been renamed “high risk” (H) in the new ATA guideline and now covers patients with MEN2A and RET codon C634 mutations. ATA-B mutations, including mutations at codons 609, 611, 618, 620 (exon 10) and 630, carry a decreased risk for severe MTC mutations (exon 11). ATA-A mutations are associated with the lowest risk. When they have preventive thyroidectomy at age 4 years, these patients have lower serum calcitonin levels, a lower tumor stage, and a better rate of biochemical cure compared to ATA-B mutation carriers of the same age. RET mutations at codons 768, 790, 791 (exon 13), 804 (exon 14), and 891 (exon 15) are seen in ATA-A mutations. The revised ATA guideline combines the current A and B levels into a new category, “moderate risk” (MOD), which includes patients with hMTC and RET codon mutations other than M918T and C634 (Tables 1 and 2) [12, 58].

Mutation locationExonMutationPhenotypeMutation risk level according to 2009ATA
Extra cellular cadherin like domain5G321RFMTC/MEN2AA
Extra cellular cysteine rich domain8C515SFMTC/MEN2AA
532 duplicationFMTCA
529/531FMTC?
G533CFMTC/MEN2AA
532 duplicationFMTCA
531/9bp duplicationFMTC/MEN2AA
10R600QFMTC/MEN2AA
K603EFMTC/MEN2AA
K603QFMTCA
Y606CFMTCA
C609F/R/G/S/YFMTC/MEN2A/HSCRB
C611R/G/F/S/W/YFMTC/MEN2A/HSCRB
C618R/G/F/S/YFMTC/MEN2A/HSCRB
C620R/G/F/S/W/YFMTC/MEN2A/HSCRB
C630R/F/S/YFMTC/MEN2AB
11D631YFMTCB
633MEN2A?
633/9bp duplicationFMTC/MEN2AB
634/12bp duplicationFMTC/MEN2AB
C634RMEN2A/CLAC
C634G/F/S/W/YFMTC/MEN2A/CLAC
634/12bp duplicationMEN2AB
635/insertion ELCR/T636PFMTC/MEN2AA
637MEN2A?
S649LFMTC/MEN2AA
K666EFMTC/MEN2AA
13E768DFMTC/MEN2A/ sMTCA
N777SFMTCA
778FMTC?
N776SFMTC/MEN2AA
781FMTC?
L790FFMTC/MEN2AA
Y791FFMTC/MEN2AA
14V804LFMTC/MEN2AA
V804MFMTC/MEN2A/ sMTCA
V804M+E805KMEN2BD
V804M+Y806CMEN2BD
G819KFMTCA
R833CFMTC/ MEN2BA
R844QFMTC/ MEN2BA
V804M/E805KMEN 2BD
V804M/Y806CMEN 2BD
804/844FMTC?
852FMTC?
Intra cellular
tyrosine kinase
domain
15R866WFMTC/MEN2AA
876FMTC?
A883FMEN2B/ sMTCD
S891AFMTC/MEN2AA
16R912PFMTC/MEN2A/ MEN2BA
M918TsMTC/MEN2BD
920sMTC/MEN2BD
922sMTC/MEN2BD
13/14V804M+V778IFMTC/MEN2AB
14/15V804M+S904CMEN2B/ MEN2AD
13/16768/919FMTC?
14/15V804M/S904CMEN 2BD

Table 1.

Genotype-phenotype correlations and risk levels for different populations of aggressive MTC according to ATA.

Family/patient No.MTC age of onsetGenderExonCodonNucleotide/amino acid (RET Mutation)Phenotype
226F11634TGC → TAC (Cys634Tyr)MEN2A
510M14804GTG → ATG (Val804Met)Sporadic
631F10611TGC → TAC (Cys611Tyr)Sporadic
713F11634TGC → CGC (Cys634Arg)Sporadic
1127F10618TGC → CGC (Cys618Arg)Sporadic
2115F11634TGC → CGC (Cys634Arg)Sporadic
2626M11630TGC → CGC (Cys630Arg)FMTC
31/121F11634TGC → CGC (Cys634Arg)Sporadic
31/221F11634TGC → CGC (Cys634Arg)MEN2A
31/321M11634TGC → CGC (Cys634Arg)Sporadic
3920M11634TGC → CGC (Cys634Arg)Sporadic
5122M11634TGC → CGC (Cys634Arg)Sporadic
5314F16918ATG → ACG (Met918Thr)MEN2B

Table 2.

Correlation between the genetic findings and the phenotype of the respective Iranian population.

Advertisement

3. Geographical pattern of RET mutations in various populations

This phenomenon, known as allelic heterogeneity, occurs when different mutations at the same locus result in the same phenotype. A diverse mutation pattern has been identified in this gene as a result of the characterization of multiple RET proto-oncogene mutations. It suggests that these mutations in distinct populations are related with a slightly diverse phenotype in various countries. Despite the identification of codon 634 of the RET proto-oncogene as a hot spot codon in the evolution of MEN2A and FMTC, the amino acid alteration at this codon is almost unique to each group. The identification of RET mutations in MTC patients was examined extensively in a number of diverse groups, which are briefly summarized in this section (Table 3) [59].

PopulationRET mutationReference
AmericanThe V804M mutation was observed in MEN2A patients frequently, rather than many other populations in which the cysteine codons have mutated.[60, 61]
AustralianThe most frequent variants were V804L, V804M, and C634R in MEN2A patients. Furthermore, the C620R, C634Y, and C634R were found in higher incidence in FMTC cases.[62, 63]
Brazilian, Chinese, Indian and MoroccanThe C634R and the C634Y were the most common mutations in MEN2A patients, respectively. Also in Brazilian FMTCs the G533C was observed frequently. This mutation is known as a rare mutation in many other countries.[64, 65, 66, 67, 68]
Czech RepublicThe C634R mutation in MEN2A patients and V804M and Y791F mutations in FMTC patients were the most had the most common occurrence.[69]
FrenchThe C634Y and C634R mutations in MEN2A, and the C618S and C620R mutations in FMTC were the most frequent.[70, 71, 72]
GermanyThe C634R and C620F mutations in MEN2A, and the E768D mutation in FMTC were more prevalent in comparison with other mutations[58, 73]
GreekThe G533C mutation in exon eight of the RET gene was observed in FMTC and sMTC patients with high prevalence. In addition, K-RAS and BRAF gene mutations in a Greek cohort of sporadic PTC and MTC carcinomas are reported.[74, 75, 76]
IranianThe C634Y and then the C634R mutations were observed MEN2A patients with the highest frequencies. In addition, the C634Y, C634W, and C630Y mutations were the most common in FMTC. In sMTC individuals, the C634R mutation occurred with the highest frequency. Interestingly, there was not any mutation in MTC patients in hot spot codon 609 (exon10) in this population.[5, 47, 48, 50, 77, 78, 79]
ItalianThe most common mutations in MEN2A were C634R, C618R, and C634Y, respectively, and in sMTC were V804M, E768D, and S891A.[80, 81]
JapaneseThe C620Y, C634R, and C634S were frequent mutations in MEN2A patients. Also in FMTC the C618Y and C630S mutations had higher incidence.[82]
KoreanThe C634Y, C634R, and C618R were the most common mutations in MEN2A. In addition, FMTC patients in this population had high C618S mutation frequency.[83, 84, 85]
MexicanThe C634Y mutation was higher in MEN2A patients than the other mutations.[83]
PortugueseThe C634R and C611Y mutations in MEN2A, and the C634R and C634Y mutations in FMTC were common. Moreover, in sMTC the V804M, L760F, and C620R mutations were observed in most of the cases Recently, two novel mutations (C515W and T636M) associated with MTC have been also identified in this population.[2, 86, 87, 88]
South AfricaThe C634S mutation had the highest frequency in MEN2A patients.[89, 90, 91]
SpanishThe most common mutations were C634R and then C634Y in MEN2A patients, while the C634R mutation in FMTC had a higher frequency. Interestingly, the predominant mutation in MEN2B cases in all evaluated populations was M918T in exon 16 of the RET proto oncogene.[92, 93, 94, 95]

Table 3.

RET mutational spectrum in different populations.

Advertisement

4. Conclusion

RET mutations do not simply determine MTC formation. These cancers likely carry mutations in additional genes, and it may be necessary to be aware of these mutations in order to consider combination therapy. This may provide new targets for the combination of RET inhibitors with other drugs that target these pathways [7]. Although a number of patients with refractory MTC have been treated with a variety of TKIs over the past few years, it is still unclear if the RET genotype of tumor cells influences clinical response to these medications [38, 54].

The thyroid cancer is the most prevalent endocrine cancer. In terms of diagnosis and preventive treatment, MTC has the strongest hereditary component among other kinds of thyroid cancer, according to ATA standards. MEN2B RET proto-oncogene mutations appear to be predominantly fixed at the M918T location in exon 16. However, mutations associated with MEN2A and FMTC vary amongst populations. For diagnosis, it will be important to evaluate and identify population-specific trends in point mutations [12]. Although there are several approaches in the treatment of RET-associated cancers; including monoclonal antibodies, kinase inhibitors, adaptor-protein binding inhibitors, dimerization inhibitors and gene therapy. Searching for specific inhibitors of RET kinase is a promising strategy. Indeed, reagents such as antioxidants, which abrogate RET dimerization, may also be useful in the treatment of MTC and PTC. Moreover, recent advances in RNA interference technology are providing a novel tool for cancer therapy [23].

More research is required and comprehensive clinical studies must be undertaken, but the preliminary findings are encouraging and optimistic. In the fight against cancer, the in-depth study of cancer and the identification of solid therapeutic targets and effective pharmacological agents have once again proven fruitful.

References

  1. 1. Dvořáková Š, Vaclavikova E, Sýkorová V, Dušková J, Vlček P, Ryška A, et al. New multiple somatic mutations in the RET proto-oncogene associated with a sporadic medullary thyroid carcinoma. Thyroid. 2006;16(3):311-316
  2. 2. Moura MM, Cavaco BM, Pinto AE, Domingues R, Santos JR, Cid MO, et al. Correlation of RET somatic mutations with clinicopathological features in sporadic medullary thyroid carcinomas. British Journal of Cancer. 2009;100(11):1777-1783
  3. 3. Elisei R, Cosci B, Romei C, Bottici V, Renzini G, Molinaro E, et al. Prognostic significance of somatic RET oncogene mutations in sporadic medullary thyroid cancer: a 10-year follow-up study. The Journal of Clinical Endocrinology and Metabolism. 2008;93(3):682-687
  4. 4. Sippel RS, Kunnimalaiyaan M, Chen H. Current management of medullary thyroid cancer. The Oncologist. 2008;13(5):539-547
  5. 5. Hedayati M, Zarif Yeganeh M, Sheikhol Eslami S, Rezghi Barez S, Hoghooghi Rad L, Azizi F. Predominant RET germline mutations in exons 10, 11, and 16 in Iranian patients with hereditary medullary thyroid carcinoma. Journal of Thyroid Research. 2011;2011
  6. 6. Ibáñez CF. Structure and physiology of the RET receptor tyrosine kinase. Cold Spring Harbor Perspectives in Biology. 2013;5(2):a009134
  7. 7. Prazeres H, Torres J, Rodrigues F, Couto JP, Vinagre J, Sobrinho-Simoes M, et al. How to treat a signal? Current basis for RET-genotype-oriented choice of kinase inhibitors for the treatment of medullary thyroid cancer. Journal of Thyroid Research. 2011;2011
  8. 8. Agrawal N, Jiao Y, Sausen M, Leary R, Bettegowda C, Roberts NJ, et al. Exomic sequencing of medullary thyroid cancer reveals dominant and mutually exclusive oncogenic mutations in RET and RAS. The Journal of Clinical Endocrinology and Metabolism. 2013;98(2):E364-E369
  9. 9. Roman S, Lin R, Sosa JA. Prognosis of medullary thyroid carcinoma: demographic, clinical, and pathologic predictors of survival in 1252 cases. Cancer: Interdisciplinary International Journal of the American Cancer Society. 2006;107(9):2134-2142
  10. 10. De Groot JWB, Plukker JTM, Wolffenbuttel BHR, Wiggers T, Sluiter WJ, Links TP. Determinants of life expectancy in medullary thyroid cancer: age does not matter. Clinical Endocrinology. 2006;65(6):729-736
  11. 11. Pellegriti G, Leboulleux S, Baudin E, Bellon N, Scollo C, Travagli JP, et al. Long-term outcome of medullary thyroid carcinoma in patients with normal postoperative medical imaging. British Journal of Cancer. 2003;88(10):1537-1542
  12. 12. Hedayati M, Zarif Yeganeh M, Sheikholeslami S, Afsari F. Diversity of mutations in the RET proto-oncogene and its oncogenic mechanism in medullary thyroid cancer. Critical Reviews in Clinical Laboratory Sciences. 2016;53(4):217-227
  13. 13. Meng X, Lindahl M, Hyvonen ME, Parvinen M, de Rooij DG, Hess MW, et al. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science. 2000;287(5457):1489-1493
  14. 14. Hickey JG, Myers SM, Tian X, Zhu SJV, Shaw JL, Andrew SD, et al. RET-mediated gene expression pattern is affected by isoform but not oncogenic mutation. Genes, Chromosomes and Cancer. 2009;48(5):429-440
  15. 15. Arighi E, Borrello MG, Sariola H. RET tyrosine kinase signaling in development and cancer. Cytokine and Growth Factor Reviews. 2005;16(4–5):441-467
  16. 16. Colombo-Benkmann M, Li Z, Riemann B, Hengst K, Herbst H, Keuser R, et al. Characterization of the RET protooncogene transmembrane domain mutation S649L associated with nonaggressive medullary thyroid carcinoma. European Journal of Endocrinology. 2008;158(6):811-816
  17. 17. de Groot JWB, Links TP, Plukker JTM, Lips CJM, Hofstra RMW. RET as a diagnostic and therapeutic target in sporadic and hereditary endocrine tumors. Endocrine Reviews. 2006;27(5):535-560
  18. 18. Phay JE, Shah MH. Targeting RET receptor tyrosine kinase activation in cancer. Clinical Cancer Research. 2010;16(24):5936-5941
  19. 19. Airaksinen MS, Saarma M. The GDNF family: Signalling, biological functions and therapeutic value. Nature Reviews Neuroscience. 2002;3(5):383-394
  20. 20. Takahashi M. The GDNF/RET signaling pathway and human diseases. Cytokine and Growth Factor Reviews. 2001;12(4):361-373
  21. 21. Yeganeh MZ, Sheikholeslami S, Hedayati M. RET proto oncogene mutation detection and medullary thyroid carcinoma prevention. Asian Pacific Journal of Cancer Prevention. 2015;16(6):2107-2117
  22. 22. Mitsiades N, Fagin JA. Molecular genetics of thyroid cancer: pathogenetic significance and clinical applications. In: Genetic Diagnosis of Endocrine Disorders. Elsevier; 2010. pp. 117-138
  23. 23. Kodama Y, Asai N, Kawai K, Jijiwa M, Murakumo Y, Ichihara M, et al. The RET proto-oncogene: a molecular therapeutic target in thyroid cancer. Cancer Science. 2005;96(3):143-148
  24. 24. Boikos SA, Stratakis CA. Molecular mechanisms of medullary thyroid carcinoma, current approaches in diagnosis and treatment. Histology and Histopathology. 2008
  25. 25. Pützer BM, Drosten M. The RET proto-oncogene: a potential target for molecular cancer therapy. Trends in Molecular Medicine. 2004;10(7):351-357
  26. 26. Santoro M, Melillo RM, Carlomagno F, Vecchio G, Fusco A. Minireview: RET: normal and abnormal functions. Endocrinology. 2004;145(12):5448-5451
  27. 27. Encinas M, Crowder RJ, Milbrandt J, Johnson EM. Tyrosine 981, a novel ret autophosphorylation site, binds c-Src to mediate neuronal survival. Journal of Biological Chemistry. 2004;279(18):18262-18269
  28. 28. Maeda K, Murakami H, Yoshida R, Ichihara M, Abe A, Hirai M, et al. Biochemical and biological responses induced by coupling of Gab1 to phosphatidylinositol 3-kinase in RET-expressing cells. Biochemical and Biophysical Research Communications. 2004;323(1):345-354
  29. 29. Gild ML, Bullock M, Robinson BG, Clifton-Bligh R. Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nature Reviews Endocrinology. 2011;7(10):617
  30. 30. Subbiah V, Yang D, Velcheti V, Drilon A, Meric-Bernstam F. State-of-the-art strategies for targeting RET-dependent cancers. Journal of Clinical Oncology. 2020;38(11):1209
  31. 31. Cabanillas ME, Ryder M, Jimenez C. Targeted therapy for advanced thyroid cancer: Kinase inhibitors and beyond. Endocrine Reviews. 2019;40(6):1573-1604
  32. 32. Omur O, Baran Y. An update on molecular biology of thyroid cancers. Critical Reviews in Oncology/Hematology. 2014;90(3):233-252
  33. 33. Huang L, Jiang S, Shi Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). Journal of Hematology and Oncology. 2020;13(1):1-23
  34. 34. Weitzman SP, Cabanillas ME. The treatment landscape in thyroid cancer: A focus on cabozantinib. Cancer Management and Research. 2015;7:265
  35. 35. Kawamoto Y, Takeda K, Okuno Y, Yamakawa Y, Ito Y, Taguchi R, et al. Identification of RET autophosphorylation sites by mass spectrometry. Journal of Biological Chemistry. 2004;279(14):14213-14224
  36. 36. Schuringa JJ, Wojtachnio K, Hagens W, Vellenga E, Buys CHCM, Hofstra R, et al. MEN2A-RET-induced cellular transformation by activation of STAT3. Oncogene. 2001;20(38):5350-5358
  37. 37. Buckwalter TLF, Venkateswaran A, Lavender M, La Perle K, Cho J-Y, Robinson ML, et al. The roles of phosphotyrosines-294,-404, and-451 in RET/PTC1-induced thyroid tumor formation. Oncogene. 2002;21(53):8166-8172
  38. 38. Knowles PP, Murray-Rust J, Kjær S, Scott RP, Hanrahan S, Santoro M, et al. Structure and chemical inhibition of the RET tyrosine kinase domain. Journal of Biological Chemistry. 2006;281(44):33577-33587
  39. 39. Raue F, Frank-Raue K. Update multiple endocrine neoplasia type 2. Familial Cancer. 2010;9(3):449-457
  40. 40. Segouffin-Cariou C, Billaud M. Transforming ability of MEN2A-RET requires activation of the phosphatidylinositol 3-kinase/AKT signaling pathway. Journal of Biological Chemistry. 2000;275(5):3568-3576
  41. 41. Machens A, Niccoli-Sire P, Hoegel J, Frank-Raue K, van Vroonhoven TJ, Roeher H-D, et al. Early malignant progression of hereditary medullary thyroid cancer. New England Journal of Medicine. 2003;349(16):1517-1525
  42. 42. Kouvaraki MA, Shapiro SE, Perrier ND, Cote GJ, Gagel RF, Hoff AO, et al. RET proto-oncogene: a review and update of genotype–phenotype correlations in hereditary medullary thyroid cancer and associated endocrine tumors. Thyroid. 2005;15(6):531-544
  43. 43. Figlioli G, Landi S, Romei C, Elisei R, Gemignani F. Medullary thyroid carcinoma (MTC) and RET proto-oncogene: Mutation spectrum in the familial cases and a meta-analysis of studies on the sporadic form. Mutation Research/Reviews in Mutation Research. 2013;752(1):36-44
  44. 44. Strosberg JR. Update on the management of unusual neuroendocrine tumors: pheochromocytoma and paraganglioma, medullary thyroid cancer and adrenocortical carcinoma. Seminars in Oncology. 2013;40(1):120-133
  45. 45. Frank-Raue K, Rondot S, Schulze E, Raue F. Change in the spectrum of RET mutations diagnosed between 1994 and 2006. Clinical Laboratory. 2007;53(5–6):273-282
  46. 46. Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib H, et al. Medullary thyroid cancer: management guidelines of the American Thyroid Association. Thyroid. 2009;19(6):565-612
  47. 47. Alvandi E, Akrami SM, Chiani M, Hedayati M, Nayer BN, Tehrani MRM, et al. Molecular analysis of the RET proto-oncogene key exons in patients with medullary thyroid carcinoma: A comprehensive study of the Iranian population. Thyroid. 2011;21(4):373-382
  48. 48. Sheikholeslami S, Yeganeh MZ, Rad LH, Ghadaksaz HG, Hedayati M. Haplotype Frequency of G691S/S904S in the RET proto-onco-gene in Patients with medullary thyroid carcinoma. Iranian Journal of Public Health. 2014;43(2):235
  49. 49. Neocleous V, Skordis N, Portides G, Efstathiou E, Costi C, Ioannou N, et al. RET proto-oncogene mutations are restricted to codon 618 in Cypriot families with multiple endocrine neoplasia 2. Journal of Endocrinological Investigation. 2011;34(10):764-769
  50. 50. Zarif Yeganeh M, Sheikholeslami S, Dehbashi Behbahani G, Farashi S, Hedayati M. Skewed mutational spectrum of RET proto-oncogene Exon 10 in Iranian patients with medullary thyroid carcinoma. Tumor Biology. 2015;36(7):5225-5231
  51. 51. Hesanmanesh H, Zarif Yeganeh M, Sheikholeslami S, Daneshvar Farhud D, Hedayati M. Germline mutations of RET proto-oncogene exon 2 in iranian population with medullary thyroid carcinoma. Koomesh. 2016;17(3):636-642
  52. 52. Carlomagno F, Vitagliano D, Guida T, Basolo F, Castellone MD, Melillo RM, et al. Efficient inhibition of RET/papillary thyroid carcinoma oncogenic kinases by 4-amino-5-(4-chloro-phenyl)-7-(t-butyl) pyrazolo [3, 4-d] pyrimidine (PP2). The Journal of Clinical Endocrinology and Metabolism. 2003;88(4):1897-1902
  53. 53. Cuccuru G, Lanzi C, Cassinelli G, Pratesi G, Tortoreto M, Petrangolini G, et al. Cellular effects and antitumor activity of RET inhibitor RPI-1 on MEN2A-associated medullary thyroid carcinoma. Journal of the National Cancer Institute. 2004;96(13):1006-1014
  54. 54. Strock CJ, Park J-I, Rosen DM, Ruggeri B, Denmeade SR, Ball DW, et al. Activity of irinotecan and the tyrosine kinase inhibitor CEP-751 in medullary thyroid cancer. The Journal of Clinical Endocrinology and Metabolism. 2006;91(1):79-84
  55. 55. Lindsey SC, Ganly I, Palmer F, Tuttle RM. Response to initial therapy predicts clinical outcomes in medullary thyroid cancer. Thyroid. 2015;25(2):242-249
  56. 56. Wells SA Jr, Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, et al. Revised American thyroid association guidelines for the management of medullary thyroid carcinoma: The American thyroid association guidelines task force on medullary thyroid carcinoma. Thyroid. 2015;25(6):567-610
  57. 57. Tuttle RM, Haddad RI, Ball DW, Byrd D, Dickson P, Duh Q-Y, et al. Thyroid carcinoma, version 2.2014. Journal of the National Comprehensive Cancer Network. 2014;12(12):1671-1680
  58. 58. Frank-Raue K, Buhr H, Dralle H, Klar E, Senninger N, Weber T, et al. Long-term outcome in 46 gene carriers of hereditary medullary thyroid carcinoma after prophylactic thyroidectomy: Impact of individual RET genotype. European Journal of Endocrinology. 2006;155(2):229-236
  59. 59. McClellan J, King M-C. Genetic heterogeneity in human disease. Cell. 2010;141(2):210-217
  60. 60. Etit D, Faquin WC, Gaz R, Randolph G, DeLellis RA, Pilch BZ. Histopathologic and clinical features of medullary microcarcinoma and C-cell hyperplasia in prophylactic thyroidectomies for medullary carcinoma: A study of 42 cases. Archives of Pathology and Laboratory Medicine. 2008;132(11):1767-1773
  61. 61. Quayle FJ, Fialkowski EA, Benveniste R, Moley JF. Pheochromocytoma penetrance varies by RET mutation in MEN 2A. Surgery. 2007;142(6):800-805
  62. 62. Learoyd DL, Gosnell J, Elston MS, Saurine TJ, Richardson A, Delbridge LW, et al. Experience of prophylactic thyroidectomy in multiple endocrine neoplasia type 2A kindreds with RET codon 804 mutations. Clinical Endocrinology. 2005;63(6):636-641
  63. 63. Marsh DJ, Theodosopoulos G, Martin-Schulte K, Richardson A-L, Philips J, Röher H-D, et al. Genome-wide copy number imbalances identified in familial and sporadic medullary thyroid carcinoma. The Journal of Clinical Endocrinology and Metabolism. 2003;88(4):1866-1872
  64. 64. Abdelhakim A, Anne B, Mohamed K, Nadia B, Mohammed T, Taoufiq F, et al. RET genetic screening in patients with medullary thyroid cancer: The Moroccan experience. Journal of Cancer Research and Therapeutics. 2009;5(3):198
  65. 65. Benazzouz B, Hafidi A, Benkhira S, Chraibi A, Kadiri A, Hilal L. C634R mutation of the protooncongene RET and molecular diagnosis in multiple endocrine neoplasia type 2 in a large Moroccan family. Bulletin Du Cancer. 2008;95(4):457-463
  66. 66. Oliveira MNL, Hemerly JP, Bastos AU, Tamanaha R, Latini FRM, Camacho CP, et al. The RET p. G533C mutation confers predisposition to multiple endocrine neoplasia type 2A in a Brazilian kindred and is able to induce a malignant phenotype in vitro and in vivo. Thyroid. 2011;21(9):975-985
  67. 67. Sharma BP, Saranath D. RET gene mutations and polymorphisms in medullary thyroid carcinomas in Indian patients. Journal of Biosciences. 2011;36(4):603-611
  68. 68. Siqueira DR, Romitti M, da Rocha AP, Ceolin L, Meotti C, Estivalet A, et al. The RET polymorphic allele S836S is associated with early metastatic disease in patients with hereditary or sporadic medullary thyroid carcinoma. Endocrine-Related Cancer. 2010;17(4):953-963
  69. 69. Jindrichova S, Vcelak J, Vlcek P, Neradilova M, Nemec J, Bendlova B. Screening of six risk exons of the RET proto-oncogene in families with medullary thyroid carcinoma in the Czech Republic. Journal of Endocrinology. 2004;183(2):257-265
  70. 70. Chatal J-F, Campion L, Kraeber-Bodéré F, Bardet S, Vuillez J-P, Charbonnel B, et al. Survival improvement in patients with medullary thyroid carcinoma who undergo pretargeted anti-carcinoembryonic-antigen radioimmunotherapy: A collaborative study with the French Endocrine Tumor Group. J Clin Oncol. 2006;24(11):1705-1711
  71. 71. Guyétant S, Josselin N, Savagner F, Rohmer V, Michalak S, Saint-André J-P. C-cell hyperplasia and medullary thyroid carcinoma: clinicopathological and genetic correlations in 66 consecutive patients. Modern Pathology. 2003;16(8):756-763
  72. 72. Schuffeneker I, Billaud M, Calender A, Chambe B, Ginet N, Calmettes C, et al. RET proto-oncogene mutations in French MEN 2A and FMTC families. Human Molecular Genetics. 1994;3(11):1939-1943
  73. 73. Frank-Raue K, Höppner W, Frilling A, Kotzerke J, Dralle H, Haase R, et al. Mutations of the ret protooncogene in German multiple endocrine neoplasia families: relation between genotype and phenotype. German Medullary Thyroid Carcinoma Study Group. The Journal of Clinical Endocrinology and Metabolism. 1996;81(5):1780-1783
  74. 74. Goutas N, Vlachodimitropoulos D, Bouka M, Lazaris AC, Nasioulas G, Gazouli M. BRAF and K-RAS mutation in a Greek papillary and medullary thyroid carcinoma cohort. Anticancer Research. 2008;28(1A):305-308
  75. 75. Kaldrymides P, Mytakidis N, Anagnostopoulos T, Vassiliou M, Tertipi A, Zahariou M, et al. A rare RET gene exon 8 mutation is found in two Greek kindreds with familial medullary thyroid carcinoma: Implications for screening. Clinical Endocrinology. 2006;64(5):561-566
  76. 76. Sarika HL, Papathoma A, Garofalaki M, Vasileiou V, Vlassopoulou B, Anastasiou E, et al. High prevalence of exon 8 G 533 C mutation in apparently sporadic medullary thyroid carcinoma in G reece. Clinical Endocrinology. 2012;77(6):857-862
  77. 77. Ghazi AA, Bagheri M, Tabibi A, Sarvghadi F, Abdi H, Hedayati M, et al. Multiple endocrine neoplasia type 2A in an Iranian family: clinical and genetic studies. Archives of Iranian Medicine. 2014;17(5):0
  78. 78. Hedayati M, Nabipour I, Rezaei-Ghaleh N, Azizi F. Germline RET mutations in exons 10 and 11: an Iranian survey of 57 medullary thyroid carcinoma cases. Medical Journal of Malaysia. 2006;61(5):564
  79. 79. Majidi M, Haghpanah V, Hedayati M, Khashayar P, Mohajeri-Tehrani MR, Larijani B. A family presenting with multiple endocrine neoplasia type 2B: A case report. Journal of Medical Case Reports. 2011;5(1):1-3
  80. 80. Romei C, Mariotti S, Fugazzola L, Taccaliti A, Pacini F, Opocher G, et al. Multiple endocrine neoplasia type 2 syndromes (MEN 2): results from the Ita MEN network analysis on the prevalence of different genotypes and phenotypes. European Journal of Endocrinology. 2010;163(2):301-308
  81. 81. Shifrin AL, Ogilvie JB, Stang MT, Fay AM, Kuo Y-H, Matulewicz T, et al. Single nucleotide polymorphisms act as modifiers and correlate with the development of medullary and simultaneous medullary/papillary thyroid carcinomas in 2 large, non-related families with the RET V804M proto-oncogene mutation. Surgery. 2010;148(6):1274-1281
  82. 82. Kameyama K, Okinaga H, Takami H. RET oncogene mutations in 75 cases of familial medullary thyroid carcinoma in Japan. Biomedicine and Pharmacotherapy. 2004;58(6–7):345-347
  83. 83. González-Yebra B, Medrano ME, Mantilla A, Palma V, Colin C, Hernández DM, et al. Penetrance of inherited medullary thyroid carcinoma and genotype-phenotype correlation in a large multiple endocrine neoplasia type 2A family with C634Y RET mutatiom. Endocrine Pathology. 2003;14(1):71-80
  84. 84. Jae Chung Y, Kim H-H, Kim H-J, Min Y-K, Lee M-S, Lee M-K, et al. RET proto-oncogene mutations are restricted to codon 634 and 618 in Korean families with multiple endocrine neoplasia 2A. Thyroid. 2004;14(10):813-818
  85. 85. Jung J, Uchino S, Lee Y, Park H. A Korean family of familial medullary thyroid cancer with Cys618Ser RET germline mutation. Journal of Korean Medical Science. 2010;25(2):226-229
  86. 86. Bugalho MJ, Domingues R, Santos JR, Catarino AL, Sobrinho L. Mutation analysis of the RET proto-oncogene and early thyroidectomy: Results of a Portuguese cancer centre. Surgery. 2007;141(1):90-95
  87. 87. Prazeres HJ, Rodrigues F, Figueiredo P, Naidenov P, Soares P, Bugalho MJ, et al. Occurrence of the Cys611Tyr mutation and a novel Arg 886Trp substitution in the RET proto-oncogene in multiple endocrine neoplasia type 2 families and sporadic medullary thyroid carcinoma cases originating from the central region of Portugal. Clinical Endocrinology. 2006;64(6):659-666
  88. 88. Silva AL, Carmo F, Moura MM, Domingues R, Espadinha C, Leite V, et al. Identification and characterization of two novel germline RET variants associated with medullary thyroid carcinoma. Endocrine. 2015;49(2):366-372
  89. 89. Moore SW, Zaahl MG. Multiple endocrine neoplasia syndromes, children, Hirschsprung’s disease and RET. Pediatric Surgery International. 2008;24(5):521-530
  90. 90. Moore SW, Zaahl MG. Chasing the ubiquitous RET proto-oncogene in South African MEN2 families-implications for the surgeon: oncology. South African Journal of Surgery. 2010;48(4):127-131
  91. 91. Moore SW, Appfelstaedt J, Zaahl MG. Familial medullary carcinoma prevention, risk evaluation, and RET in children of families with MEN2. Journal of Pediatric Surgery. 2007;42(2):326-332
  92. 92. Belli S, Storani ME, Dourisboure RJ, Podestá EJ, Solano AR. Study of RET protooncogene in multiple endocrine neoplasm 2A and in familial medullary thyroid carcinoma. Clinical pathological findings in asymptomatic carriers. Medicina. 2003;63(1):41-45
  93. 93. Milos IN, Frank-Raue K, Wohllk N, Maia AL, Pusiol E, Patocs A, et al. Age-related neoplastic risk profiles and penetrance estimations in multiple endocrine neoplasia type 2A caused by germ line RET Cys634Trp (TGC> TGG) mutation. Endocrine-Related Cancer. 2008;15(4):1035-1041
  94. 94. Nelson Wohllk G, Youlton R, Cote GJ, Gagel RF. Germline mutations of the ret proto-oncogen in Chilean patients with hereditary and sporadic medullary thyroid carcinoma. Rev. Méd. Chile. 2001;129(7)
  95. 95. Sobrino PS, Fernández CP, Gil PG, Gil BM, Pedrosa AP, Carballeira RP, et al. Phenotype of the C634Y mutation in the RET proto-oncogene in MEN2A: Report of a family. Endocrinología y Nutrición (English Edition). 2011;58(5):229-235

Written By

Shabnam Heydarzadeh and Mehdi Hedayati

Submitted: 16 July 2022 Reviewed: 08 November 2022 Published: 17 April 2023