ERC
HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
 QUICK SEARCH:   [advanced]


     


Endocrine-Related Cancer 15 (4) 871 -884     DOI: 10.1677/ERC-08-0098
Copyright © 2008 by the Society for Endocrinology
This Article
Right arrow Abstract Freely available
Right arrow Full Text (PDF)
Right arrow Alert me when this article is cited
Right arrow Alert me if a correction is posted
Services
Right arrow Similar articles in this journal
Right arrow Similar articles in PubMed
Right arrow Alert me to new issues of the journal
Right arrow Download to citation manager
Citing Articles
Right arrow Citing Articles via HighWire
Right arrow Citing Articles via Web of Science (3)
Right arrow Citing Articles via Google Scholar
Google Scholar
Right arrow Articles by Sakorafas, G. H
Right arrow Articles by Peros, G.
Right arrow Search for Related Content
PubMed
Right arrow PubMed Citation
Right arrow Articles by Sakorafas, G. H
Right arrow Articles by Peros, G.

The genetic basis of hereditary medullary thyroid cancer: clinical implications for the surgeon, with a particular emphasis on the role of prophylactic thyroidectomy

George H Sakorafas, Helmut Friess1 and George Peros

From the 4th Department of Surgery, Athens University Medical School, ATTIKON University Hospital, Arkadias 19-21, 115 26 Athens, Greece1 Department of Surgery, Technische Universitt Mnchen, Munich, Germany

(Correspondence should be addressed to G H Sakorafas; Email: georgesakorafas{at}yahoo.com)


    Abstract
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
Medullary thyroid cancer (MTC) may occur either sporadically or on a hereditary basis. Hereditary MTC may be observed with either multiple endocrine neoplasia syndromes (MEN 2A and MEN 2B) or as familial MTC (FMTC). Despite the rarity of these syndromes, early diagnosis is especially important, since MTC is a lethal disease if not promptly and appropriately treated. Recently, the development of genetic testing and direct DNA analysis allows the identification of asymptomatic patients. Surgical prophylaxis should be considered in these cases, ideally to prevent the development of MTC. During the recent decade, the concept of ‘codon-directed’ timing of prophylactic surgery emerged as a reasonable strategy in the management of these patients. Currently, genetic analysis offers the possibility to define genotype-phenotype correlations and to adjust the time of prophylactic surgery. Hereditary MTC is a model of genetically determined cancer in which both diagnostic and therapeutic strategies rely on the identification of specific mutations.


    Introduction
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
Medullary thyroid cancer (MTC) is a relatively rare type of thyroid cancer, constituting 4-8% of all thyroid malignancies (Poston 1993, Phay et al. 2000, Clayman & El-Baradie 2003). MTC was first described by Hazard et al. (1959). It arises from the parafollicular C-cells, thereby differing from the classic follicular cell origin of tumors of the thyroid, and may occur either sporadically or, less commonly, on a hereditary basis (Phay et al. 2000, Clayman & El-Baradie 2003). The familial occurrence of MTC was first reported in 1961, when MTC was diagnosed in a pair of young siblings (in their early 20s) and whose mother had previously died following surgery for thyroid carcinoma (Friedell et al. 1961). Since that time, it has been observed that the familial MTC is inherited as an autosomal dominant trait in about 20-30% of patients (Komminoth et al. 1995, Phay et al. 2000, Brandi et al. 2001, Clayman & El-Baradie 2003, Shaha et al. 2006). Sipple (1961) reported the association of thyroid cancer with pheochromocytoma. Williams (1963), at the Hammersmith Hospital, was the first to point out that the pattern of thyroid cancer in this disease was always medullary. The genetic component of the disease was first described by Schmimke & Hartmann (1965) who demonstrated the mode of inheritance through an autosomal dominant trait. In 1968, Steiner et al. (1968) coined the term multiple endocrine neoplasia type 2 (MEN 2) and included hyperparathyroidism as part of the constellation of clinical problems. In 1977, Norman noted that, apart from the dominant pattern of familial disease, there may be a recessive inheritance in isolated families (Normann 1977).

MTC is the most common cause of death in patients with MEN 2 (Modigliani et al. 1998, Skinner et al. 2005). Once established, it spreads early on and poses many difficulties in clinical management, since it is both chemo- and radioresistant. Early surgery is the only therapeutic method that can achieve cure of the patient. In the past, the diagnosis in the familial setting of MTC was generally not made until the disease was symptomatic or clinically evident. Thus, the MTC was not discovered until the third or fourth decade of life in affected patients and was usually at an advanced and incurable stage (Skinner 2003). Recent advances in molecular biology allowed an in-depth understanding of the mechanisms of carcinogenesis and the detection of specific genetic alterations associated with MTC. These advances have led to major changes in the diagnostic and therapeutic algorithm in these increasingly common malignancies (You et al. 2007). The concept of prophylactic surgery (thyroidectomy) has proven effective in preventing or curing hereditary MTC in young patients. The aim of this paper is to present currently available data regarding the molecular basis of hereditary MTC and to emphasize the role of prophylactic surgery in these patients.


    Pathology, epidemiology, clinical, and genetic aspects of the hereditary MTC
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
MTC is a rare calcitonin-producing tumor of the parafollicular or C-cells of the thyroid gland (Kakudo et al. 1985). Multifocal C-cell hyperplasia (CCH) is a precursor lesion to hereditary MTC. The progression from CCH to microscopic MTC is undoubtedly variable and may take several years (Papotti et al. 1993). Metastasis may be in the central and lateral, cervical, and mediastinal lymph nodes or more distantly in the lung, liver, or bone.

Hereditary MTC occurs with either multiple endocrine neoplasia type 2A (in 60% of cases), MEN type 2B (5%), or familial medullary thyroid carcinoma (FMTC, 35%; Brandi et al. 2001). To avoid delayed diagnosis and adverse clinical outcomes, the clinician should always consider the possibility of hereditary MTC; therefore, care must be taken in labeling a patient as a ‘sporadic’ MTC and then failing to screen the rest of the family carefully. The astute physician should keep in his/her mind the possibility of an underlying MEN 2 syndrome in these cases, and therefore he/she should screen for pheochromocytoma and hypercalcemia preoperatively and during follow-up.

MEN2 syndromes

Multiple endocrine neoplasia type 2 (MEN 2) is an autosomal dominant inherited disorder consisting of two main different syndrome variants, the MEN 2A and MEN 2B syndromes. These two MEN 2 variants have the greatest syndromic consistency (Brandi et al. 2001). Other clinical variants have also been described (Table 1; Verdy et al. 1982, Farndon et al. 1986, Donovan et al. 1989, Nunziata et al. 1989). As a result of the high penetrance of the MEN 2 phenotype, tumors may present at an earlier age, even under the age of 5 years (Machens et al. 2001). Especially, medullary thyroid cancer (MTC) may have a more aggressive biological behavior (Schilling et al. 2001). In MEN 2B families, MTC can be observed even within the first year of life in germline mutation carriers (Sanso et al. 2005), while metastases have been reported at 3 years of age (Kaufman et al. 1982).


View this table:
[in this window]
[in a new window]

 
Table 1 Typical RET and variations

 
MEN2A
MEN 2A accounts for the vast majority (up to 90%) of MEN 2A cases and is characterized by MTC (in nearly 100% of patients), pheochromocytoma (in about 50% of cases), and primary hyperparathyroidism (in about 30% of patients; Vasen et al. 1987, Howe et al. 1993, Lee & Norton 2000, Skinner 2003, Danko & Skinner 2006). The MTC is the first neoplasm to develop, and, as noted above, it represents the most common cause of cancer-related deaths in MEN patients (Danko & Skinner 2006).

MEN 2B
MEN 2B is very rare and presents as MTC and bilateral pheochromocytomas, but without any parathyroid gland abnormalities (Howe et al. 1993, Ponder & Smith 1996, Skinner 2003). MEN 2B is also associated with distinct physical manifestations and an easily recognizable phenotype (marfanoid habitus, multiple mucosal neuromas of the lips and tongue, intestinal ganglioneuromatosis, and pigment spots on the face (Carney et al. 1976, O'Riordain et al. 1995, Lee & Norton 2000, Skinner 2003). The MTC associated with MEN 2B is extremely aggressive than in the other familial syndromes, often developing metastases in early childhood (Samaan et al. 1991, Skinner et al. 1996). As a result, most patients die before the age of 30 years. Diarrhea from humoral factors produced by MTC combined with gastrointestinal dysmotility from intestinal ganglioneuromatosis can significantly alter quality of life (Brandi et al. 2001).

FMTC

In FMTC, there are no manifestations of the disease outside of the thyroid gland and MTC is the only neoplasm present (Farndon et al. 1986, Skinner 2003). It is an autosomal dominant inherited disease, in general less aggressive, with a late onset of the disease clinically between 30 and 50 years of age (Niccoli-Sire et al. 2003).

Genetic alterations in hereditary MTC

In 1988, linkage analysis was applied to kindreds affected with MEN 2A and a mutation was found on chromosome 10q11.2. In 1993 and 1994, germline mutations in the RET (rearranged during transfection) proto-oncogene, located at 10q11.2, were identified in patients with MEN 2A, MEN 2B or FMTC (Donis-Keller et al. 1993, Mulligan et al. 1993, Carlson et al. 1994, Eng et al. 1994, Hofstra et al. 1994). The encoded RET protein, a receptor tyrosine kinase expressed in neural crest-derived cells (i.e., thyroid parafollicular cells, parathyroid cells, adrenal medulla chromaffin cells, and enteric autonomic plexuses), mediates downstream pathways of cell survival and mitogenesis (Frilling et al. 2003, Shaha et al. 2006). Specific germ line mutations in the gene lead to constitutive activation of the receptor and cause the dominant inherited cancer MEN 2 syndrome and FMTC (Skinner 2003, Gertner & Kebebew 2004; Table 2). Because RET is a proto-oncogene, a single activating mutation of one allele is sufficient to cause neoplastic transformation. A mutation in the RET proto-oncogene is present in a variable proportion (usually around 25%) of cases with MTC (Leboulleux et al. 2004). MTC became the first identified inherited human neoplasm to be associated with the dominant activation of a proto-oncogene. Recent advances in our understanding of molecular carcinogenesis of MTC have significant clinical implications and, nowadays, genetic screening is considered of key importance in the management of these patients (Moore et al. 2007). Identification of a germline mutation in a patient with MTC enables classification of the tumor as being of the inherited type, regardless of the absence of family history or associated endocrinopathies (Bugalho et al. 2007). Currently, DNA-based strategies allow a correct identification of asymptomatic carriers, while avoiding the unpleasant stimulation tests.


View this table:
[in this window]
[in a new window]

 
Table 2 RET mutations associated with RET syndromes

 
In 98% of MEN 2A patients and in 80-90% of FMTC patients, the germinal RET mutation occurs in one of five cysteine codons of the extracellular domain of the RET protein: 609, 611, 618, 629 (exon 10), and 634 (exon 11; Eng et al. 1996). In the remaining cases, cysteine codon mutations may occur in codon 610, 620, or 630 (Komminoth et al. 1995, Bugalho et al. 2007). These ‘gain of function’ mutations result in receptor dimerization and constitutive activation, ultimately giving rise to MEN 2A and FMTC (Chappuis-Flament et al. 1998, Gimm et al. 2004, You et al. 2007). Codon 634 (exon 11) is the most commonly affected (mutated in 80% of MEN 2A cases; Machens et al. 2003a,b).

In addition to these genetic changes, other rarer non-cysteine mutations located within the intracellular catalytic domain of RET have been described. These mutations can give rise to FMTC (codons 768, 790, 791, 804, 806, and 891) and to MEN 2B (codons 833 and 918) (Bolino et al. 1995, Eng et al. 1995a,b, Hofstra et al. 1997, Berndt et al. 1998, You et al. 2007). More than 95% of patients with MEN 2B have a mutation at codon 918, causing receptor autophosphorylation and activation (Hansford & Mulligan 2000). Altered signaling is responsible for the more aggressive MTC and the visible phenotypic features associated with MEN 2B (Santoro et al. 1995, Hansford & Mulligan 2000). Rarely, more than one mutation in RET can be associated with MTC.

Of particular clinical importance is that these mutations are associated with specific component tumors and phenotypes (Eng 1999). In other words, a significant genotype-phenotype interaction exists with some mutations being significantly less aggressive than others (Yip et al. 2003, Gertner & Kebebew 2004, Kouvaraki et al. 2005). No RET genetic variations appear to carry the same risk, however, and the quantification of specific risk of RET - related genetic variations appear to be codon specific. Machens et al. (2005) have proposed the stratification of the risk (high versus intermediate versus low risk) based on the specific encountered mutations ((codons 634 and 918) versus (codons 790, 620 and 611) versus (codons 768 and 804) respectively; see below, Management). Codon 634 and 918 mutations have the highest transforming activity and correlate strongly with the onset of MEN 2. As a consequence, there is a significant correlation between codon 634 mutation and early development of MTC, pheochromocytoma, and hyperparathyroidism (Calender et al. 1997, Ponder 1999, Szinnai et al. 2003). Codon 634 mutations have been associated with the development of MTC as early as at the age of 15 months (Modigliani et al. 1998) and with the development of cervical lymph node metastases at the age of 5 years and 11 months (Gill et al. 1996). Other cysteine codon mutations (i.e., codons 609, 618, and 620) have a lower risk (intermediate risk group). They are associated with a 3x-5x risk of MTC, but only half the risk of a 634 mutation (Ito et al. 1997). Mutations in non-cysteine domains of the RET (see above) afford a weaker activation, resulting in a attenuated form with low penetrance, late-onset MTC (classically between 30 and 50 years of age) and the FMTC phenotype only and infrequently result in disease-associated mortality (Bolino et al. 1995, Pasini et al. 1997, Rossel et al. 1997, Berndt 1998, Feldman et al. 2000, Lombardo et al. 2002, Niccoli-Sire et al. 2003, Leboulleux et al. 2004, Menacho et al. 2005, Vestergaard et al. 2007). This has led to the supposition that these patients have a mild ‘C-cell disease phenotype’ (Berndt et al. 1998, Fattoruso et al. 1998). A possible explanation of these differences seen between the codons is that the mechanism of RET activation may differ between specific codons within the gene. For example, mutations at residues 618 and 620 (and 609 to a much lesser extent) result in a much lower expression of the cell-surface form of RET than the 634 mutation (Ito et al. 1997, Pasini et al. 1997, Chappuis-Flament et al. 1998).

This knowledge generated the concept of ‘codon-directed’ timing of surgery (see ‘Management’ below); according to this surgical philosophy, the timing of prophylactic surgery is dictated by the specific genotype (i.e., surgery being performed before the earliest reported age of MTC diagnosis for that specific codon). However, the clinician should keep in his/her mind that - although generally the risk is codon specific - the age of onset of MTC is not always constant and that there are always exceptions to the rule. For example, in some cases, these mutations have been associated with a more aggressive biological behavior (development of locally invasive disease and distant metastases) (Fattoruso et al. 1998, Feldman et al. 2000, Niccoli-Sire et al. 2001, 2003). It may be possible that, in association with these genetic changes, there may be some additional influence by other modifying factors (Cebrian et al. 2005). Therefore, the risk associated with some (especially non-cysteine) mutations may be uncertain.


    Diagnostic approach
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
Although MEN 2 syndromes are rare, early diagnosis is especially important. Up until the last decade, these syndromes were recognized mainly through family history. Nowadays, with the development of genetic testing and direct DNA analysis, molecular techniques are increasingly used to identify asymptomatic patients. As a result of these recent advances, MEN 2 syndromes are increasingly and more commonly diagnosed in the pediatric population, presenting challenging clinical scenarios. Surgical prophylaxis has a central role in the management of these patients.

Measurement of basal and stimulated calcitonin levels

Calcitonin is the primary secretory product of malignant C-cells of MTC; it can be measured by using a sensitive immunometric calcitonin assay (Barbot et al. 1994). Calcitonin is important as an excellent tumor marker (Gagel et al. 1988, Pacini et al. 1994). Calcitonin levels (both basal as well as stimulated by pentagastrin, calcium, or both; Heshmati et al. 1997, Niccoli et al. 1997, Cohen et al. 2000) are nearly always elevated with MTC.

Basal calcitonin levels

MTC can be diagnosed before being clinically evident (presymptomatic diagnosis) based on increased basal calcitonin levels (Melvin et al. 1971). In the study by Niccoli-Sire et al. (2003), when a basal calcitonin level was elevated, all but one patient had MTC and nodal metastases were found in 37.1% of the patients. These investigators reported a continuous progression in the staged disease with regard to basal calcitonin levels above 30 pg/ml. Nodal involvement was nearly constant when calcitonin was more than 100 pg/ml and was associated with macroscopic MTC in one half of the cases (Niccoli-Sire et al. 2003). It therefore appears that progression from CCH parallels increasing basal calcitonin levels and more advanced staged disease (Niccoli-Sire et al. 1999, 2001, Cohen et al. 2000); in other words, there is a positive correlation between tumoral mass and calcitonin levels. However, despite that basal calcitonin levels are higher in MTC than in CCH, this difference cannot be used for an accurate differential diagnosis between these two entities (Franck-Raue et al. 1996, Guyetant et al. 1997, Feldman et al. 2000, Gimm et al. 2002, Niccoli-Sire et al. 2003).

Stimulated calcitonin levels

The sensitivity of biochemical diagnosis of MTC and MEN 2 syndromes was further increased by the use of secretagogues (i.e., pentagastrin or calcium) to stimulate tumor secretion of calcitonin (Niccoli-Sire et al. 2003). In this test, pentagastrin is infused intravenously (0.5 µg/kg, for 3 min) to stimulate secretion of calcitonin, and blood samples are collected before and 3, 5, and 10 min after the initiation of the pentagastrin injection. It should, however, be noted that pentagastrin peptide for parenteral (i.v.) testing is currently in very restricted availability. Alternatively, calcium gluconate can be used as a secretagogue (2 mg/kg infused over 1 min) (Skinner 2003). Calcitonin response is considered normal (i.e., negative pentagastrin test results) when less than 10 pg/ml (Barbot et al. 1994). However, this upper limit obviously is assay dependent. Pentagastrin test allows the detection of MTC at an early stage - even with normal basal calcitonin levels, when MTC is isolated to the thyroid gland, thereby increasing the chances of cure by surgery (Wells et al. 1982). Pentagastrin test can accurately detect C-cell disease. For example, it has been shown that when preoperative peak calcitonin level was pathologic, MTC was present in over 92% of cases (Heizmann et al. 2006). However, up to 20% of patients had normal pentagastrin-stimulated calcitonin levels, although CCH or MTC was documented on histologic examination (Dralle et al. 1998, Niccoli-Sire et al. 1999). Most authors agree that differential diagnosis between CCH and MTC is not possible based on stimulated calcitonin levels (Niccoli-Sire 2003, Heizmann et al. 2006).

A major limitation of biochemical diagnosis of MTC is that the diagnosis is made after the development of the disease (even if diagnosed before the appearance of any clinical signs or symptoms of MTC); at this stage, a truly prophylactic surgery is not possible. Moreover, measurement of calcitonin levels (basal and stimulated (which is unpleasant to the patient)) needs to be done yearly and indefinitely (Ponder et al. 1988).

Today the role of basal and stimulated calcitonin levels is of limited value in accurately predicting the stage of neoplastic development of the C-cells or the long-term patient outcome (Iler et al. 1999). These tests should not be used to define the timing of thyroid surgery in children with RET syndromes (see below, management; Iler et al. 1999). Today, they have been replaced by genetic analysis that allows the identification of mutations in the RET proto-oncogene (see below).

Genetic testing

Screening of DNA for the detection of RET mutations is effective and widely available (Skinner et al. 2005). When performed rigorously, it reveals a RET mutation in over 95% of MEN 2 index cases (Brandi et al. 2001). The RET test has a higher rate of true positives and lower rates of false negatives and false positives than the calcitonin tests. It allows early diagnosis of MEN 2 syndromes, before the development of any neoplastic changes within the thyroid, thereby facilitating surgical decision making regarding prophylactic thyroidectomy (Skinner 2003). Genetic testing is useful for confirming hereditary MTC and for identifying asymptomatic gene carriers in known kindreds with MTC who probably will develop thyroid cancer (Lips et al. 1994, Wells et al. 1994). This strategy has obvious clinical implications. Early genetic screening in children of parents with MEN 2 or FMTC has led to prophylactic total thyroidectomy. Interestingly, genetic analysis offers the possibility to define genotype-phenotype correlations and to adjust the time of prophylactic surgery (Machens et al. 2005). Hereditary MTC is thus a model of genetically determined cancer in which both diagnostic and therapeutic strategies rely on the identification of specific mutations (see below).


    Management
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
In older MEN 2A series, with treatment initiated after the identification of a thyroid nodule, MTC progressed and showed a 15-20% cancer mortality (Kakudo et al. 1985). Early thyroidectomy reduced mortality from hereditary MTC to less than 5% (Brandi et al. 2001). Before the recognition of MEN 2 sudden death from pheochromocytoma was frequent in these families, perhaps as frequent as death from progression of MTC (Steiner et al. 1968). However, it is probable that improved management of pheochromocytoma has decreased the rate of premature mortality in MEN 2 even more than has the improved management of MTC (Brandi et al. 2001).

The specifically mutated codon of RET correlates with the MEN 2 variant, including the aggressiveness of MTC. Thus, the mutated RET codon and the features within the family should receive careful attention in planning thyroid management. The MTC risk can be stratified, according to the known RET mutation (Brandi et al. 2001).

In patients with RET positivity a clear progression from CCH to MTC has been documented (Machens et al. 2003a,b, Ashworth 2005). This observation raised the question about the role of preventive measures in RET mutation carriers. Obviously, the first step in the management of these patients is the recognition of these genetic changes; RET genetic screening is therefore indicated. Interestingly, the American Society of Clinical Oncology (ASCO) and-recently-the National Comprehensive Cancer Network proposed that patients with apparently sporadic MTC should undergo RET mutational analysis to rule out germline mutations, which would prompt the testing of offspring for similar mutations (Statement of the ASCO 1996, NCCN Oncology Practice Guidelines 2005). Early identification of mutation carriers can significantly change the decision making in these patients. As above noted, in MEN 2A, the risk of developing MTC is 90-100%, and therefore the only curative treatment is total thyroidectomy, ideally before the development of MTC, or at least when the MTC is still confined within the thyroid and before the spread of the disease beyond the gland (Wells et al. 1994, Gimm et al. 2001, Danko & Skinner 2006, Heizmann et al. 2006, You et al. 2007). To appreciate the value of prophylactic thyroidectomy, the clinician should keep in his/her mind that, although infrequently, even microscopic MTC may be aggressive and can metastasize in about 5% of patients (Guyetant et al. 1999). Some large series have confirmed this hypothesis, as all individuals who underwent thyroidectomy had MTC or its assumed precursor lesion CCH (Lips et al. 1994, Dralle et al. 1998, Niccoli-Sire et al. 1999). This therapeutic strategy is especially important since advanced disease is incurable by conventional chemotherapy or radiotherapy, which further emphasizes the importance of early detection and prompt intervention (Niccoli-Sire et al. 1999, Machens et al. 2003a,b, Skinner et al. 2005, You et al. 2007). Thyroidectomy performed on the basis of positive genetic screening and before the development of MTC is truly prophylactic, while thyroidectomy performed after the development of MTC is essentially therapeutic. RET mutation carriers should also be screened for pheochromocytoma (Heizmann et al. 2006). Historically, the MEN 2 syndromes represent the first disease for which prophylactic removal of an organ target is recommended before the development of malignancy, solely on the basis of a genetic test (Skinner 2003). It has been, in fact, the near-perfect model for developing such approaches in that, unlike all other familial cancers, there exists an accurate diagnostic genetic test, there is minimal morbidity from the surgical intervention of total thyroidectomy, organ function is able to be fully replaced with thyroxine, and a test of subsequent disease status (calcitonin) is available (Gosnell et al. 2006).

The likelihood of a RET germline mutation in a patient with apparently sporadic MTC is 1-7% (Eng et al. 1995a,b). A RET germline mutation is more likely if apparently sporadic MTC has an early age of onset, if there is multiplicity within the thyroid, or if it is observed in association with a pheochromocytoma (Brandi et al. 2001).

Obviously, surgical morbidity should be minimal and within acceptable limits. The reported incidences of permanent hypocalcemia and recurrent laryngeal nerve dysfunction have ranged from 1 to 7% and 1% respectively (Dralle et al. 1998, Niccoli-Sire et al. 1999, Brandi et al. 2001, Ukkat et al. 2001, Rodriguez et al. 2002). While in situ preservation of parathyroid tissue is often possible, long-term success has been demonstrated with parathyroid autotransplantation in either the brachioradialis or the sternocleidomastoid muscle (Olson et al. 1996). Certainly, the risk of complications following thyroidectomy is decreased when the procedure is performed by surgeons with considerable experience in the procedure (Sosa et al. 1998). Anesthesia and airway management in children may be viewed as a barrier, but the risk is decreased when a pediatric anesthesiologist is available.

Timing of prophylactic thyroidectomy

Contrasting with the remarkable achievements in genetic diagnosis, no major progress has been registered concerning treatment, particularly for distant metastases. As above noted, children carrying activating RET mutations firstly develop thyroid CCH, which progressively transforms into MTC, in virtually all gene carriers and often before the age of 5 years (Machens et al. 2002, Piolat et al. 2006). MTC then rapidly spreads to cervical lymph nodes and ultimately leads to distant metastasis, the main cause of death of these patients. Operation removing the target organ (thyroidectomy), ideally before the development of MTC, remains the only effective preventive/therapeutic approach (Evans et al. 1999, Gimm et al. 2001), thus explaining the importance of early diagnosis of germline carriers as well as of early prophylactic thyroidectomy (Niccoli-Sire et al. 1999, Machens et al. 2003a,b, Skinner et al. 2005). Often, prophylactic thyroidectomy in RET mutation carriers should be performed before age of 5 years or in the first decade of life (Brandi et al. 2001, Moore et al. 2007).

The limitations of measurements of calcitonin levels (basal/stimulated) are well-known (see above; Iler et al. 1999). Surgery on the basis of abnormal calcitonin tests fails to be truly prophylactic, since a positive pentagastrin test result indicates the presence of C-cell disease or even MTC (Machens et al. 2001, Niccoli-Sire 2003). Genetic testing has a higher rate of true positives and lower rates of false negatives and false positives than the calcitonin tests, and it facilitates earlier thyroidectomy (Brandi et al. 2001) and currently prophylactic thyroidectomy should be carried out regardless of serum calcitonin levels (Gosnell et al. 2006). At the MEN97 Workshop consensus was reached that the decision to perform thyroidectomy in MEN 2 should be based predominately on the result of RET mutation testing rather than on calcitonin testing (Lips 1998). Nowadays, it is well known that the onset of neoplastic development and the aggressiveness of MTC are closely related to the location of the mutated codon within the RET gene. Thus, genotype analysis allows the classification of carriers of the RET mutation into risk categories and a more tailored approach to management of these patients (Shapiro et al. 2003). Thus, thyroidectomy can be performed before the development of MTC, thereby preventing this potentially lethal disease by removing the still ‘normal’ but at-risk thyroid parenchyma. The finding of normal pathology in RET mutation carriers has been reported elsewhere and supports the concept that early recognition and treatment can in fact lead to surgical cure before the development of CCH or MTC (Kebebew et al. 1999). Recommendations for the optimal timing of prophylactic surgery depend on this risk stratification (Brandi 2001).

The International RET Mutation Consortium correlated codon genotypes with clinical aggressiveness of hereditary MTC and provided guidelines for the timing of prophylactic thyroidectomy (Eng et al. 1996, Brandi 2001). Patients with MEN 2B or RET mutations in codons 883, 918, or 922 are at the highest risk (level 3) and should undergo thyroidectomy within the first year of life. Others have suggested prophylactic thyroidectomy even earlier (within the first 6 months or preferentially within the first month) in these cases (You et al. 2007). To support their recommendations, these investigators emphasized that the finding of microscopic MTC within the first year of life in this setting is common, and that even metastasis during the first year of life has been described (Stjernholm et al. 1980, Kaufman et al. 1982, Samaan et al. 1991, Smith et al. 1999).

Patients with MEN 2A or FMTC who have mutations in codons 611, 618, 620, and 634 are at high risk (level2) and thyroidectomy should be performed before the age of 5 years (Wells et al. 1994, Brandi 2001, Machens et al. 2001, Danko & Skinner 2006, You et al. 2007). However, early thyroidectomy with a codon 634 mutation has helped to identify microscopic MTC in a child as young as 2 years of age (Modigliani et al. 1998).

Children with RET codon 609, 768, 790, 791, 804, and 891 mutations or with FMTC are classified as level 1 (the lowest risk among the three RET codon mutation stratification categories; Brandi et al. 2001). The biological behavior of MTC in patients with these mutations is variable, but in general MTC grows more slowly and develops at a later age than with the high risk mutations. However, lymph node metastases and death caused by MTC have been observed for mutation in each of these except codons 790 and 791. Currently, there is no clear consensus regarding the optimal timing of thyroidectomy in this group of patients. Some recommend surgery before age of 5 years (as in the high risk (level 2) group of patients) and others suggest surgery before age of 10 years, and still others believe that intervention is only required when stimulated calcitonin levels are abnormal (Brandi et al. 2001, Colombo-Benkmann et al. 2002, Fitze et al. 2002, Vestergaard et al. 2007). In this group of patients, if delay of thyroidectomy is decided, a clear plan of evaluation should be followed (You et al. 2007).

It appears that this surgical philosophy is effective in preventing the development of MTC and improves the long-term results of treatment (Skinner et al. 2005). Indeed, Szinnai et al. (2003) found a difference between patients who underwent ‘early’ thyroidectomy (by age 1-5 years) and those with ‘late’ thyroidectomy (by age 6-20 years). Whereas all children with early total thyroidectomy seemed to be cured (median follow-up, 3 years; maximum, 9 years), the patients aged between 6 and 20 years at operation showed a considerable proportion of persistent or recurrent disease (55%) after 10 years. Skinner et al. (2005) reported the long-term outcome of prophylactic thyroidectomy in 50 children (mean age 10 years) with allelic mutations characteristic of MEN 2A. These patients had prophylactic thyroidectomy, central lymphadenectomy, and routine parathyroid autotransplantation. CCH, microscopic MTC, or macroscopic MTC was found in 94% of the patients and nodal metastases were present in 6%. After a minimum follow-up of 5 years, 88% of the 50 patients remained disease-free, as defined by undetectable calcitonin levels following provocative testing. Other institutional series with smaller numbers of patients (Lallier et al. 1998, van Heurn et al. 1999, Sanso et al. 2005), less complete or shorted follow-up (Lips et al. 1994, Pacini et al. 1995, Dralle et al. 1998, Niccoli-Sire et al. 1999, Ukkat et al. 2001, Rodriguez et al. 2002) have reported variably defined ‘cure rates’ of 76-100%. As expected, the prognosis of hereditary MTC in more advanced stages is not favorable. When hereditary MTC is detected clinically (as palpable tumors), survival is significantly diminished (5- and 10-year survival rates, 78 and 64% respectively (Bergholm et al. 1990).

Not surprisingly, a significant delay between the date of genetic diagnosis and the date of thyroidectomy has been reported (Piolat et al. 2006). Mostly, this delay was related to the time parents needed to accept and allow for a total thyroidectomy in their asymptomatic and apparently ‘healthy’ child. Cultural factors may also affect the acceptance of early prophylactic surgery in asymptomatic children.

The role of cervical lymph node dissection

It is evident that if total thyroidectomy is truly prophylactic (i.e., it is performed before the development of MTC), then the at-risk thyroid parenchyma is removed and the risk of MTC is totally (at least theoretically) eliminated. In this case, prophylactic cervical lymph node dissection is not indicated. The problem is that none can be sure preoperatively that no MTC will be found after a presumably prophylactic thyroidectomy in mutation carriers. Therefore, the role of cervical lymph node dissection during prophylactic thyroidectomy remains controversial. Lymph node metastases are common in MTC and, albeit rarely, can be observed even in the presence of microscopic MTC (Dralle et al. 1998). MTC size cannot be used to predict the presence of cervical lymph node metastases. Lymph node metastases in patients with hereditary MTC of less than 1 cm may reach an incidence of 20% (Bigner et al. 1981, Dralle et al. 1995). Recurrent disease is observed in a large proportion of patients (33%) if a central neck dissection is omitted at the time of total thyroidectomy for established MTC (Decker 1992). These data emphasize the significance of the unresolved problem about the role of cervical lymph node dissection in the management of patients at risk for the development of hereditary MTC. The radicality during initial surgery is of utmost importance, because of its influence on the outcome of patients with hereditary MTC (Ellenhom et al. 1993, Kallinowski et al. 1993, Dralle et al. 1995, Tamagnini et al. 2005).

The role of cervical lymph node dissection during prophylactic thyroidectomy remains controversial. In the past, patients who underwent prophylactic thyroidectomy did not undergo concomitant standard lymphadenectomy (Dralle et al. 1995). Currently, it is generally accepted that in MEN 2B patients undergoing prophylactic thyroidectomy, central lymph node dissection is indicated and, if nodal metastases are found, a more extensive node dissection should be considered (Brandi et al. 2001). In contrast, despite that nodal metastases from MTC have been reported in a child at age of 5 years (Gill et al. 1996), there is no consensus regarding the need for prophylactic dissection of central lymph nodes in MEN 2A and FMTC, with most authors supporting prophylactic thyroidectomy alone (i.e., without concomitant cervical lymph node dissection), since in these patients the likelihood of metastatic lymph node disease is very low (Lallier et al. 1998, Brandi et al. 2001, Frilling et al. 2003, Machens et al. 2003a,b, Skinner 2003, Danko & Skinner 2006, Gosnell et al. 2006, Butter et al. 2007). In the presence of elevated basal or stimulated calcitonin levels (which indicates the presence of C-cell disease, either CCH or MTC), central lymph node dissection at the time of total thyroidectomy should be considered (Brandi et al. 2001, Niccoli-Sire et al. 2003, Heizmann et al. 2006). Obviously, a more aggressive neck dissection should be performed if there is evidence of involved lymph nodes in the lateral neck. Cervical lymph node dissection should also be performed in older (above 10 years) patients with MEN 2A and FMTC undergoing, for a variety of reasons, delayed thyroidectomy or when MTC is diagnosed during a presumably prophylactic thyroidectomy (Heizmann et al. 2006).

From a technical point of view, it should be noted that central lymph node dissection during the primary operative procedure should be preferred because the risk of permanent complications increases if lymphadenectomy is carried out as a reoperation (Colombo-Benkmann et al. 1998). However, primary central lymph node dissection is associated with a higher rate of permanent hypoparathyroidism and recurrent laryngeal nerve damage compared with total thyroidectomy alone (Piolat et al. 2006). To remove potentially existing locoregional metastases from MTC, it is recommended that the lymph nodes be removed in the central neck from the hyoid bone to the clavicles, and between the carotid sheaths (Skinner 2003). This is usually the first nodal basin to collect metastatic MTC.

Complications, follow-up and recurrence of the disease

Currently, prophylactic surgery can be performed safely and with minimal postoperative morbidity (Iler et al. 1999). Expertise is required from the part of both the surgeon and the anesthesiologist to maintain complications rates within acceptable limits. In experienced hands, the risks associated with a delayed thyroidectomy outweigh the morbidity of early surgery (Iler et al. 1999).

Postoperatively, follow-up is indicated to allow early detection of a possible recurrence. As expected, recurrence is more common in node-positive patients (Skinner et al. 2005). Calcitonin is an excellent tumor marker not only for diagnostic but also for follow-up purposes (Shaha et al. 2006). Generally, elevated calcitonin levels after surgery are generally the first sign of persistent or recurrent disease (Brandi et al. 2001, Shaha et al. 2006). Stimulated calcitonin levels have also been used to detect recurrent disease (Skinner 2003). Unfortunately, due to the lack of published data, it remains unknown how frequently these tests should be performed. Furthermore, it is unknown how many years should pass without the development of MTC before the child can be declared ‘cured’ by his thyroidectomy.

If the basal or stimulated plasma calcitonin levels are high after primary thyroid surgery, the presence and extent of local and/or distant (metastatic) disease should be documented (Dottorini et al. 1996, Scopsi et al. 1996, Modigliani et al. 1998). Unfortunately, as a result of the exquisite sensitivity of calcitonin levels in detecting MTC, localization of recurrent metastatic disease may be very challenging (Danko & Skinner 2006). Most radiographic modalities have very low sensitivities for the detection of early recurrent or metastatic MTC. Following adequate diagnosis of recurrence, a decision regarding optimal management should be made (Dottorini et al. 1996, Scopsi et al. 1996, Bergholm et al. 1997). If there is no evidence of distant metastases and if local disease is found or suspected in the neck and/or upper mediastinum, then reoperation is advocated. Exploration of the mediastinum is controversial because of the greater morbidity and the few examples of cures. If distant metastases are found, surgery is not indicated.


    Declaration of interest
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
There is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.


    Funding
 
This scientific work did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.


    References
 Top
 Abstract
 Introduction
 Pathology, epidemiology,...
 Diagnostic approach
 Management
 Declaration of interest
 References
 
Ashworth M 2005 The pathology of preclinical medullary thyroid carcinoma. Endocrine Pathology 15 227-232.

Barbot N, Calmettes C, Schuffenecker I, Saint-Andre JP, Franc B, Rohmer V, Jallet P & Bigorgne JC 1994 Pentagastrin stimulation test and early diagnosis of MTC using an immunoradiometric assay of calcitonin: comparison with genetic screening in hereditary MTC. Journal of Clinical Endocrinology and Metabolism 78 114-120.[Abstract]

Bergholm U, Adami HO, Bergstrom R, Backdahl M & Akerstrom G 1990 Long-term survival in sporadic and familial medullary thyroid carcinoma with special reference fo clinical characteristics as prognostic factors. The Swedish MTC Study Group. Acta Chirurgica Scandinavica 156 37-46.[Web of Science][Medline]

Bergholm U, Bergstrom R & Ekbom A 1997 Long-term follow-up of patients with medullary carcinoma of the thyroid. Cancer 79 132-138.[CrossRef][Web of Science][Medline]

Berndt I, Reuter M, Saller B, Frank-Rauer K, Groth P, Grussendorf M, Rauer F, Ritter MM & Hoppner W 1998 A new hot spot for mutations in the ret proto-oncogene causing familial medullary thyroid carcinoma and multiple endocrine neoplasia type 2A. Journal of Clinical Endocrinology and Metabolism 83 770-774.[Abstract/Free Full Text]

Bigner SH, Cox EB, Mendelsohn G, Baylin SB, Wells SA Jr & Eggleston JC 1981 Medullary carcinoma of the thyroid in the multiple endocrine neoplasia IIA syndrome. American Journal of Surgical Pathology 5 459-472.[Medline]

Bolino A, Schuffenecker I, Luo Y, Seri M, Silengo M, Tocco T, Chabrier G, Houdent C, Mourat A & Schlumberger M 1995 RET mutations in exons 13 and 14 of FMTC patients. Oncogene 10 2415-2419.[Web of Science][Medline]

Brandi ML, Gagel RF, Angeli A, Bilezikian JP, Beck-Peccoz P, Bordi C, Conte-Devolx B, Falchetti A, Gheri RG & Libroia A 2001 Guidelines for diagnosis and therapy of MEN type 1 and type 2. Journal of Clinical Endocrinology and Metabolism 86 5658-5671.[Abstract/Free Full Text]

Bugalho MJ, Domingues R, Santos JR, Catarino AL & Sobrinho L 2007 Mutation analysis of the RET proto-oncogene and early thyroidectomy: results of a Portuguese cancer centre. Surgery 141 90-95.[CrossRef][Web of Science][Medline]

Butter A, Gagne J, Al-Jazaeri A, Emran MA, Deal C & St-Vil D 2007 Prophylactic thyroidectomy in pediatric carriers of multiple endocrine neoplasia type 2A or familial medullary thyroid carcinoma: mutation in C620 is associated with Hirschprung's disease. Journal of Pediatric Surgery 42 203-206.[Medline]

Calender A, Giraud S, Schuffenecker I, Lenoir GM, Gaudray P, Courseaux A, Porchet N, Aubert JP & Zhang CX 1997 Genetic testing in presympomatic diagnosis of multiple endocrine neoplasia. Hormone Research 47 199-210.[Medline]

Carlson KM, Dou S, Chi D, Scavarda N, Toshima K, Jackson CE, Wells SA Jr, Goodfellow PJ & Donis-Keller H 1994 Single missense mutation in the tyrosine kinase catalytic domain of the RET protooncogene is associated with multiple endocrine neoplasia type 2B. PNAS 91 1579-1583.[Abstract/Free Full Text]

Carney JA, Go VL, Sizemore GW & Hayles AB 1976 Alimentary tract ganglioneuromatosis. A major component of the syndrome of multiple endocrine neoplasia, type 2 B. New England Journal of Medicine 295 1287-1291.[Web of Science][Medline]

Cebrian A, Lesueur F, Martin S, Leyland J, Ahmed S, Luccarini C, Smith PL, Luben R, Whittaker J & Pharoah PD 2005 Polymorphisms in the initiators of RET signalling pathway and susceptibility to sporadic medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 90 6268-6274.[Abstract/Free Full Text]

Chappuis-Flament S, Pasini A, De Vita G, Segouffin-Cariou C, Fusco A, Attie T, Lenoir GM, Santoro M & Billaud M 1998 Dual effect on the RET receptor of MEN 2 mutations affecting specific extracytoplasmic cysteines. Oncogene 17 2851-2861.[CrossRef][Web of Science][Medline]

Clayman GL & El-Baradie TS 2003 Medullary thyroid cancer. Otolaryngologic Clinics of North America 36 91-105.[CrossRef][Medline]

Cohen R, Campos JM, Salaun C, Heshmati HM, Proye C, Sarfati E, Henry JF, Niccoli-Sire P & Modigliani E 2000 Preoperative calcitonin levels are predictive of tumor size and postoperative calcitonin normalization in medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 85 919-922.[Abstract]

Colombo-Benkmann M, Raff J, Raue F, Klar E & Herfarth C 1998 Effect of primary surgical therapy on the course of C-cell carcinoma of the thyroid gland. Langenbeck's Archives of Surgery 115 1041-1043.

Colombo-Benkmann M, Bramswig J, Hoppner W, Gellner R, Hengst K, Bocker W & Senninger N 2002 Surgical strategy in a kindred with a rare RET protooncogene mutation of variable penetrance with regard to multiple endocrine neoplasia. World Journal of Surgery 26 1286-1290.[CrossRef][Medline]

Danko ME & Skinner MA 2006 Surgical intervention in children with MEN type 2. Current Opinion in Pediatrics 18 312-315.[Medline]

Decker RA 1992 Long-term follow-up of a large North American kindred with multiple endocrine neoplasia type 2A. Surgery 112 1066-1072.[Medline]

Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K, Lairmore TC, Howe JR, Moley JF, Goodfellow P & Wells SE Jr 1993 Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC. Human Molecular Genetics 2 851-856.[Abstract/Free Full Text]

Donovan DT, Levy ML, Furst EJ, Alford BR, Wheeler T, Tschen JA & Gagel RF 1989 Familial cutaneous lichen amyloidosis in association with multiple endocrine neoplasia type 2A: a new variant. Henry Ford Hospital Medical Journal 37 147-150.[Medline]

Dottorini ME, Assi A, Sironi M, Sangali G, Spreafico G & Colombo L 1996 Multivariate analysis of patients with medullary thyroid carcinoma. Prognostic significance and impact on treatment of clinical and pathological variables. Cancer 77 1556-1565.

Dralle H, Scheumann GF, Proye C, Bacourt F, Frilling A, Limbert F, Gheri G, Henry JF, Berner M, Niederle B et al. 1995 The value of lymph node dissection in hereditary medullary thyroid carcinoma: a retrospective, European, multicentric study. Journal of Internal Medicine 238 357-361.[Medline]

Dralle H, Gimm O, Simon D, Frank-Raue K, Gortz G, Niederle B, Wahl RA, Koch B, Walgenbach S, Hampel R et al. 1998 Prophylactic thyroidectomy in 75 children and adolescents with hereditary medullary thyroid carcinoma: German and Austrian experience. World Journal of Surgery 22 744-750.[CrossRef][Web of Science][Medline]

Ellenhom JD, Shah JP & Brennan MF 1993 Impact of therapeutic regional lymph node dissection for medullary carcinoma of the thyroid gland. Surgery 114 1078-1082.[Web of Science][Medline]

Eng C 1999 RET in the development of human cancer. Journal of Clinical Oncology 17 380-393.[Abstract/Free Full Text]

Eng C, Smith DP, Mulligan LM, Nagai MA, Healey CS, Ponder MA, Gardner E, Scheumann GF, Jackson CE, Tunnacliffe A et al. 1994 Point mutation within the tyrosine kinase domain of the RET proto-oncogene in multiple endocrine neoplasia type 2B and related sporadic tumors. Human Molecular Genetics 3 237-241.[Abstract/Free Full Text]

Eng C, Smith DP, Mulligan LM, Healey CS, Zvelebil MJ, Stonehouse TJ, Ponder MA, Jackson CE, Waterfield MD & Ponder BA 1995a A novel point mutation in the tyrosine kinase domain of the RET proto-oncogene in sporadic medullary thyroid carcinoma and in a family with FMTC. Oncogene 10 509-513.[Web of Science][Medline]

Eng C, Mulligan LM, Smith DP, Healey CS, Frilling A, Raue F, Newmann HP, Ponder MA & Ponder BA 1995b Low frequency of germline mutations in the RET proto-oncogene in patients with apparently sporadic medullary thyroid carcinoma. Clinical Endocrinology 43 123-127.[Medline]

Eng C, Clayton D, Schuffenecker I, Lenoir G, Cote G, Gagel RF, van Amstel HK, Lips CJ, Nishisho I, Takai SI et al. 1996 The relationship between specific RET proto-oncogene mutations and disease phenotype in MEN type 2. International RET mutation consortium analysis. Journal of the American Medical Association 276 1575-1579.[Abstract/Free Full Text]

Evans DB, Fleming JB, Lee JE, Cote G & Gagel RF 1999 The surgical treatment of medullary thyroid carcinoma. Seminars in Surgical Oncology 16 50-63.[CrossRef][Medline]

Farndon JR, Leight GS, Dilley WG, Baylin SB, Smallridge RC, Harrison TS & Wells SA Jr 1986 Familial medullary thyroid carcinoma without associated endocrinopathies: a distinct clinical entity. British Journal of Surgery 73 278-281.[Web of Science][Medline]

Fattoruso O, Quadro L, Libroia A, Verga U, Lupoli G, Cascone E & Colantuoni V 1998 A GTG to ATG novel point mutation at codon 804 in exon 14 of the RET proto-oncogene in two families affected by familial medullary thyroid carcinoma. Human Mutation 1 S167-S171.[Medline]

Feldman GL, Edmonds MW, Ainsworth PJ, Schuffenecker I, Lenoir GM, Saxe AW, Talpos GB, Roberson J, Petrucelli N & Jackson CE 2000 Variable expressivity of familial medullary thyroid carcinoma due to a RET V804M (GTG->ATG) mutation. Surgery 128 93-98.[CrossRef][Web of Science][Medline]

Fitze G, Schierz M, Bredow J, Saeger HD, Roesner D & Schackert HK 2002 Various penetrance of famiial medullary thyroid carcinoma in patients with RET protooncogene codon 790/791 germline mutations. Annals of Surgery 236 570-575.[CrossRef][Web of Science][Medline]

Franck-Raue K, Hoppner W, Frilling A, Kotzerke J, Dralle H, Haase R, Mann K, Seif F, Kirchner R, Rendl J et al. 1996 Mutations of the RET protooncogene in german MEN families: relation between genotype and phenotype. Journal of Clinical Endocrinology and Metabolism 81 1780-1783.[Abstract]

Friedell GH, Carey RJ & Rosen H 1961 Familial thyroid cancer. Cancer 15 241-245.

Frilling A, Weber F, Tecklenborg C & Broelsch CE 2003 Prophylactic thyroidectomy in multiple endocrine neoplasia: the impact of molecular mechanisms of RET proto-oncogene. Langenbeck's Archives of Surgery 388 17-26.[Medline]

Gagel RF, Tashjian AH Jr, Cumming T, Papathanasopoulos N, Kaplan MM, DeLellis RA, Wolfe HJ & Reichlin S 1988 The clinical outcome of prospective screening for MEN type 2A: a 18-year experience. New England Journal of Medicine 318 478-484.[Web of Science][Medline]

Gertner ME & Kebebew E 2004 Multiple endocrine neoplasia type 2. Current Treatment Options in Oncology 5 315-325.[CrossRef][Medline]

Gill JR, Reyes-Mugica M, Iyengar S, Kidd KK, Touloukian RJ, Smith C, Keller MS & Genel M 1996 Early presentation of metastatic medullary carcinoma in multiple endocrine neoplasia, type IIA. Implications for therapy. Journal of Pediatrics 129 459-464.[CrossRef][Web of Science][Medline]

Gimm O, Sutter T & Dralle H 2001 Diagnosis and therapy of sporadic and familial medullary thyroid carcinoma. Journal of Cancer Research and Clinical Oncology 127 156-165.[CrossRef][Web of Science][Medline]

Gimm O, Niederle BE, Weber T, Bockhorn M, Ukkat J, Brauckhoff M, Thanh PN, Frilling A, Klar E, Niederle B et al. 2002 RET proto-oncogene mutations affecting codon 790/791: a mild form of MEN type 2A syndrome? Surgery 132 952-959.[CrossRef][Web of Science][Medline]

Gimm O, Ukkat J, Niederle BE, Weber T, Thanh PN, Brauckhoff M, Niederle B & Dralle H 2004 Timing and extent of surgery in patients with familial medullary thyroid carcinoma/multiple endocrine neoplasia 2A - related RET mutations not affecting codon 634. World Journal of Surgery 28 1312-1326.[CrossRef][Web of Science][Medline]

Gosnell JE, Sywak MS, Sidhu SB, Gough IR, Learoyd DL, Robinson BG & Delbridge LW 2006 New era: prophylactic surgery for patients with MEN-2A. Australian and New Zealand Journal of Surgery 76 586-590.

Guyetant S, Rousselet MC, Durigon M, Chappard D, Frank B, Guerin O & Saint-Andre JP 1997 Sex related C-cell hyperplasia in the normal human thyroid: a quantitative autopsy study. Journal of Clinical Endocrinology and Metabolism 82 42-47.[Abstract/Free Full Text]

Guyetant S, Dupre F, Bigorgne JC, Franc B, Dutrieux-Berger N, Lecomte-Houcke M, Patey M, Caillou B, Viennet G, Guerin O et al. 1999 Medullary thyroid microcarcinoma: a clinicopathologic retrospective study of 38 patients with no prior familial disease. Human Pathology 30 957-963.[CrossRef][Web of Science][Medline]

Hansford JR & Mulligan LM 2000 Multiple endocrine neoplasia type 2 and RET: from neoplasia to neurogenesis. Journal of Medical Genetics 37 817-827.[Abstract/Free Full Text]

Hazard JB, Hawk WH & Crile G 1959 Medullary (solid) carcinoma of the thyroid. A clinico-pathologic entity. Journal of Clinical Endocrinology 19 152-155.

Heizmann O, Haecker FM, Zumsteg U, Muller B, Oberholzer M & Oertli D 2006 Presymptomatic thyroidectomy in MEN type 2A. European Journal of Surgical Oncology 32 98-102.[Medline]

Heshmati HM, Ghabib H, van Heerden JA & Sizemore GW 1997 Advances and controversies in the diagnosis and management of medullary thyroid carcinoma. American Journal of Medicine 103 60-69.[CrossRef][Web of Science][Medline]

van Heurn LW, Schaap C, Sie G, Haagen AA, Gerver WJ, Freling G, van Amstel HK & Heineman E 1999 Predictive DANN testing for multiple endocrine neoplasia 2: a therapeutic challenge of prophylactic thyroidectomy in very young children. Journal of Pediatric Surgery 34 568-571.[CrossRef][Web of Science][Medline]

Hofstra RMW, Landsvater RM, Ceccherini I, Stupl RP, Stelwagen T, Luo Y, Pasini B, Hoppener JW, van Amstel HK, Romeo G et al. 1994 A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature 367 375-376.[CrossRef][Medline]

Hofstra RM, Fattoruso O, Quadro L, Wu Y, Libroia A, Verga U, Colantuoni V & Buys CH 1997 A novel point mutation in the intracellular domain the ret proto-oncogene in a family with medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 82 4176-4178.[Abstract/Free Full Text]

Howe JR, Norton JA & Wells SA Jr 1993 Prevalence of pheochromocytoma and hyperparathyroidism in multiple endocrine neoplasia type 2A: results of long-term follow-up. Surgery 114 1070-1077.[Web of Science][Medline]

Iler MA, King DR, Ginne-Pease ME, O'Dorisio TM & Sotos JF 1999 Multiple endocrine neoplasia type 2A: a 25-year review. Journal of Pediatric Surgery 34 92-97.[CrossRef][Web of Science][Medline]

Ito S, Iwashita T, Asai N, Murakami H, Iwata Y, Sobue G & Takahashi M 1997 Biological properties of RET with cysteine mutations correlate with multiple endocrine neoplasia type 2A, gamilial medullary thyroid carcinoma, and Hirschprung's disease phenotype. Cancer Research 57 2870-2872.[Abstract/Free Full Text]

Kakudo K, Carney JA & Sizemore GW 1985 Medullary carcinoma of the thyroid. Biologic behavior of the sporadic and familial neoplasm. Cancer 55 2818-2821.

Kallinowski F, Buhr HJ, Meybier H, Eberhardt M & Herfarth C 1993 Medullary carcinoma of the thyroid - therapeutic strategy derived from fifteen years of experience. Surgery 114 491-496.[Medline]

Kaufman FR, Roe TF & Isaacs H Jr 1982 Metastatic medullary thyroid carcinoma in young children with mucosan neuroma syndrome. Pediatrics 70 263-267.[Abstract/Free Full Text]

Kebebew E, Tresler PA, Siperstein AE, Duh OY & Clark OH 1999 Normal thyroid pathology in patients undergoing thyroidectomy for finding a RETgene germline mutation: a report of three cases and review of the literature. Thyroid 9 127-131.[Web of Science][Medline]

Komminoth P, Kunz EK, Matias-Guiu X, Hiort O, Christiansen G, Colomer A, Roth J & Heitz PU 1995 Analysis of RET protooncogene point mutations distinguishes heritable from non-heritable medullary thyroid carcinomas. Cancer 76 479-489.

Kouvaraki MA, Shapiro SE, Perrier ND, Cote GJ, Gagel RF, Hoff AO, Sherman SI, Lee JE & Evans DB 2005 RET protooncogene: a review and update of genotype-phenotype correlations in hereditary medullary thyroid cancer and associated endocrine tumors. Thyroid 15 531-544.[CrossRef][Web of Science][Medline]

Lallier M, St-Vil D, Giroux M, Huot C, Gaboury L, Oligny L & Desjardins JC 1998 Prophylactic thyroidectomy for medullary thyroid carcinoma in gene carriers of MEN2 syndrome. Journal of Pediatric Surgery 33 846-848.[CrossRef][Web of Science][Medline]

Leboulleux S, Baudin E, Travagli JP & Schlumberger M 2004 Medullary thyroid carcinoma. Clinical Endocrinology 61 299-310.[CrossRef][Medline]

Lee NC & Norton JA 2000 Multiple endocrine neoplasia type 2 B - genetic basis and clinical expression. Surgical Oncology 9 111-118.[CrossRef][Web of Science][Medline]

Lips CJM 1998 Clinical management of the MEN syndromes: results of a computerized opinion poll at the 6th International Workshop on Multiple Endocrine Neoplasia and von-Hippel-Lindau disease. Journal of Internal Medicine 243 589-594.[CrossRef][Medline]

Lips CJ, Landsvater RM, Hoppener JW, Geerdink RA, Blijham G, van Veen JM, van Gils AP, de Wit MJ, Zewald RA, Berends MJ et al. 1994 Clinical screening as compared with DNA analysis in families with MEN type 2A. New England Journal of Medicine 331 828-835.[CrossRef][Web of Science][Medline]

Lombardo F, Baudin E, Chiefari E, Arturi F, Bardet S, Cailou B, Conte C, Dallapicolla B, Giuffrida D, Bidart JM et al. 2002 Familial medullary thyroid carcinoma: clinical variability and low aggressiveness associated with RET mutation at codon 804. Journal of Clinical Endocrinology and Metabolism 87 1674-1680.[Abstract/Free Full Text]

Machens A, Gimm O, Hinz R, Hoppner W, Boehm BO & Dralle H 2001 Genotype-phenotype correlations in hereditary medullary thyroid carcinoma: oncological features and biochemical properties. Journal of Clinical Endocrinology 86 1104-1109.

Machens A, Niccoli-Sire P, Hoegel J, Frank-Raue K, van Vroonhoven TJ, Roeher HD, Wahl RA, Lamesch P, Raue F, Conte-Devolx B et al. 2003a Early malignant progression of hereditary medullary thyroid cancer. New England Journal of Medicine 349 1517-1525.[CrossRef][Web of Science][Medline]

Machens A, Holzhausen HJ, Thanh PN & Dralle H 2003b Malignant progression from C-cell hyperplasia to medullary thyroid carcinoma in 167 carriers of RET germline mutations. Surgery 134 425-431.[CrossRef][Web of Science][Medline]

Machens A, Ukkat J, Brauckhoff M, Gimme O & Dralle H 2005 Advances in the management of hereditary medullary thyroid cancer. Journal of Internal Medicine 257 50-59.[CrossRef][Web of Science][Medline]

Melvin KE, Miller HH & Tashijan AH Jr 1971 Early diagnosis of medullary carcinoma of the thyroid gland by means fo calcitonin assay. New England Journal of Medicine 285 1115-1120.[Web of Science][Medline]

Menacho IP, Koster R, van der Sloot AM, Quax WJ, Osinga J, van der Sluis T, Hollema H, Burzynski GM, Gimm O, Buys CH et al. 2005 RET - familial medullary thyroid carcinoma mutants Y791F and S891A activate a Src/JAK/STAT3 pathway, independent of glial cell line-derived neurotrophic factor. Cancer Research 65 1729-1737.[Abstract/Free Full Text]

Modigliani E, Cohen R, Campos JM, Conte-Devolx B, Maes B, Boneu A, Schlumberger M, Bigorgne JC, Dumontier P, Leclerc L et al. 1998 Prognostic factors for survival and for biochemical cure in medullary thyroid carcinoma: results in 899 patients. The GETC Study Group. Clinical Endocrinology 48 265-273.[CrossRef][Medline]

Moore SW, Appfelstaedt J & Zaahl MG 2007 Familial medullary carcinoma prevention, risk evaluation, and RET in children of families with MEN 2. Journal of Pediatric Surgery 42 326-332.[Medline]

Mulligan LM, Kwok JBJ, Healey CS, Elsdon MJ, Eng C, Gardner E, Love DR, Mole SE, Moore JK, Papi L et al. 1993 Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature 363 458-460.[CrossRef][Medline]

NCCN Oncology Practice Guidelines 2005 (V.1.2005), www.nccn.org/physician_gls/_guidelines.html.

Niccoli P, Wion-Barbot N, Caron P, Henry JF, de Micco C, Saint Andre JP, Bigorgne JC, Modigliani E & Conte-Devolx B 1997 Interest of routine measurement of serum calcitonin: study in a large series of thyroidectomized patients. Journal of Clinical Endocrinology and Metabolism 82 338-341.[Abstract/Free Full Text]

Niccoli-Sire P, Murat A, Baudin E, Henry JF, Proye C, Bigorgne JC, Bstandig B, Modigliani E, Morange S, Schlumberger M et al. 1999 Early or prophylactic thyroidectomy in MEN2/FMTC gene carriers: results in 71 thyroidectomized patients. European Journal of Endocrinology 141 468-474.[Abstract]

Niccoli-Sire P, Murat A, Rohmer V, Franc S, Charbier G, Baldet L, Maes B, Savagner F, Giraud S, Bezieau S et al. 2001 Familial medullary thyroid carcinoma (FMTC) with non cysteine RET mutations: study in a large series of patients. Journal of Clinical Endocrinology and Metabolism 86 3746-3753.[Abstract/Free Full Text]

Niccoli-Sire P, Murat A, Rohmer V, Gibelin H, Chabrier G, Conte-Devolx B, Visset J, Ronceray J, Jaeck D, Henry JF et al. 2003 When should thyroidectomy be performed in familial medullary thyroid carcinoma gene carriers with non-cysteine RET mutations? Surgery 134 1029-1036.[CrossRef][Web of Science][Medline]

Normann T 1977 Medullary thyroid carcinoma in Norway: epidemiology and genetic data. Acta Pathologica et Microbiologica Scandinavica 85 775-786.

Nunziata V, Giannattasio R, di Giovanni G, D'Armiento MR & Mancini M 1989 Herditary localized prutitus in affected members of a kindred with multiple endocrine neoplasia type 2A (Sipple's syndrome). Clinical Endocrinology 30 57-63.[Medline]

Olson JA Jr, DeBenedetti MK, Baumann DS & Wells SA Jr 1996 Parathyroid autotransplantation during thyroidectomy. Results of long-term follow-up. Annals of Surgery 223 472-480.[CrossRef][Web of Science][Medline]

O'Riordain DS, O'Brien T, Crotty TB, Gharib H, Grant CS & van Heerden JA 1995 Multiple endocrine neoplasia type 2B: more than an endocrine disorder. Surgery 118 936-942.[CrossRef][Web of Science][Medline]

Pacini F, Fontanelli M, Fugazzola L, Elisei R, Romei C, DiCoscio G, Miccoli P & Pinchera A 1994 Routine measurement of secum calcitonin in nodular thyroid disease allows the preoperative diagnosis of unsuspected sporadic medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 78 826-829.[Abstract]

Pacini F, Romei C, Miccoli P, Elisei R, Molinaro E, Mancusi F, Iacconi P, Basolo F, Martino E & Pinchera A 1995 Early treatment of hereditary medullary thyroid carcinoma after attribution of multiple endocrine neoplasia type 2 gene carrier status by screening for ret gene mutations. Surgery 118 1031-1035.[CrossRef][Web of Science][Medline]

Papotti M, Botto Micca F, Favero A, Palestini N & Bussolati G 1993 Poorly differentiated thyroid carcinomas with primordial cell component. A group of aggressive lesions sharing insular trabecular and solid patterns. American Journal of Surgical Pathology 17 291-301.[Web of Science][Medline]

Pasini A, Geneste O, Legrand P, Schlumberger M, Rossel M, Fournier L, Rudkin BB, Schuffenecker I, Lenoir GM & Billaud M 1997 Oncogene activation of RET by two distinct FMTC mutations affecting the tyrosine kinase domain. Oncogene 15 393-402.[CrossRef][Web of Science][Medline]

Phay JE, Moley JF & Lairmore TC 2000 Multiple endocrine neoplasias. Seminars in Surgical Oncology 18 324-332.[CrossRef][Medline]

Piolat C, Dyon JF, Sturm N, Pinson S, Bost M, Jouk PS, Plantaz D & Chabre O 2006 Very early prophylactic thyroid surgery for infants with a mutation of the RET proto-oncogene at codon 634: evaluation of the implementation of international guidelines for MEN type 2 in a single centre. Clinical Endocrinology 65 118-124.[CrossRef][Medline]

Ponder BAThe phenotypes associated with ret mutations in the multiple endocrine neoplasia type 2 syndromeCancer Research 59 Suppl_7 1999 1736S-1741S.

Ponder BA & Smith D 1996 The MEN II syndromes and the role of the ret proto-oncogene. Advances in Cancer Research 70 179-222.[Web of Science][Medline]

Ponder BA, Ponder MA, Coffey R, Pembrey ME, Gagel RF, Telenius-Berg M, Semple P & Easton DF 1988 Risk estimation and screening in families of patients with medullary thyroid carcinoma. Lancet 1 397-401.[Web of Science][Medline]

Poston GJMedullary carcinoma of the thyroidJ Lynn & SR Bloom Surgical Endocrinology. 1993Butterworth-HeinemannOxford:270-276.

Rodriguez GJ, Balsalobre MD, Pomares F, Torregrosa NM, Carbonell P, Glower G, Sola J, Tebar J & Parrilla P 2002 Prophylactic thyroidectomy in MEN 2A syndrome: experience in a single center. Journal of the American College of Surgeons 195 159-166.[CrossRef][Medline]

Rossel M, Pasini A, Chappuis S, Geneste O, Fournier L, Schuffenecker I, Takahashi M, van Grunsven LA, Urdiales JL & Rudkin BB 1997 Distinct biological properties of two RET isoforms activated by MEN 2A and MEN 2B mutations. Oncogene 14 265-275.[CrossRef][Web of Science][Medline]

Samaan NA, Draznin MB, Halpin RE, Bloss RS, Hawikins E & Lewis RA 1991 Multiple endocrine syndrome type IIb in early childhood. Cancer 68 1832-1834.

Sanso GE, Domene HM, Garcia R, Pusiol E, De M, Roque M, Ring A, Perinetti H, Elsner B & Lorcansky S 2005 Very early detection of RET proto-oncogene mutation is crucial for preventive thyroidectomy in MEN type 2 children: presence of C-cell malignant disease in asymptomatic carriers. Cancer 94 323-330.

Santoro M, Carlomagno F, Romano A, Bottaro DP, Dathan NA, Grieco M, Fusco A, Vecchio G, Matoskova B, Kraus MH et al. 1995 Activation of RET as a dominant transforming gene by germline mutations of MEN2A and MEN2B. Science 267 381-383.[Abstract/Free Full Text]

Schilling T, Burck J, Sinn HP, Clemens A, Otto HF, Hoppner W, Herfarth C, Ziegler R, Schwab M & Raue F 2001 Prognostic value of codon 918 (ATG->ACG) RET proto-oncogene mutations in sporadic medullary thyroid carcinoma. International Journal of Cancer 95 62-66.[CrossRef][Web of Science][Medline]

Schmimke RN & Hartmann WH 1965 Familial amyloid producing medullary thyroid carcinoma and pheochromocytoma. A distinct genetic entity. Annals of Internal Medicine 95 1027-1039.

Scopsi L, Sampietro G, Boracchi P, Del BO R, Gullo M, Placucci M & Pilotti S 1996 Multivariate analysis of prognostic factors in sporadic medullary carcinoma of the thyroid. A retrospective study of 109 consecutive patients. Cancer 78 2173-2183.

Shaha AR, Cohen T, Ghossein R & Tuttle M 2006 Late-onset medullary carcinoma of the thyroid: need for genetic testing and prophylactic thyroidectomy in adult family members. Laryngoscope 116 1704-1707.[Medline]

Shapiro SE, Cote GC, Lee JE, Gagel RF & Evans DB 2003 The role of genetics in the surgical management of familial endocrinopathy syndromes. Journal of the American College of Surgeons 197 818-831.[CrossRef][Medline]

Sipple JF 1961 Association of pheochromocytoma with carcinoma of the thyroid gland. American Journal of Medicine 31 163-166.[CrossRef][Web of Science]

Skinner MA 2003 Management of hereditary thyroid cancer in children. Surgical Oncology 12 101-104.[Medline]

Skinner MA, DeBenedetti MK, Moley JF, Norton JA & Wells SA Jr 1996 Medullary thyroid carcinoma in children with multiple endocrine neoplasia types 2A and 2B. Journal of Pediatric Surgery 31 177-182.[CrossRef][Web of Science][Medline]

Skinner MA, Moley JA, Diley WG, Owzar K, Debenedetti MK & Wells SA Jr 2005 Prophylactic thyroidectomy in multiple endocrine neoplasia type 2A. New England Journal of Medicine 353 1105-1113.[CrossRef][Web of Science][Medline]

Smith VV, Eng C & Milla PJ 1999 Intestinal ganglioneuromatosis and multiple endocrine neoplasia type 2B: implications for treatment. Gut 45 143-146.[Abstract/Free Full Text]

Sosa JA, Bowman HM, Tielsch JM, Powe NR, Gordon TA & Udelsman R 1998 The importance of surgeon experience for clinical and economic outcomes from thyroidectomy. Annals of Surgery 228 320-330.[CrossRef][Web of Science][Medline]

Statement of the American Society of Clinical OncologyGenetic testing for cancer susceptibility, adopted on February 20, 1996Journal of Clinical Oncology 14 1996 1730-1736.[Abstract/Free Full Text]

Steiner AL, Goodman AD & Powers SR 1968 Study of a kindred with pheochromocytoma, hyperparathyroidism and Cushing's disease; multiple endocrine neoplasia type 2. Medicine 47 371-379.[CrossRef][Medline]

Stjernholm MR, Freudenbourg JC, Mooney HS, Kinney FJ & Deftos LJ 1980 Medullary carcinoma of the thyroid before age 2 years. Journal of Clinical Endocrinology and Metabolism 51 252-253.[Abstract/Free Full Text]

Szinnai G, Meier C, Komminoth P & Zumsteg UW 2003 Review of multiple endocrine neoplasia type 2A in children: therapeutic results of early thyroidectomy and prognostic value of codon analysis. Pediatrics 111 E132-E139.

Tamagnini P, Iacobone M, Sebag F, Marcy M, De Micco C & Henry JF 2005 Lymph node involvmenet in macroscopic medullary thyroid carcinoma. British Journal of Surgery 92 449-453.[CrossRef][Web of Science][Medline]

Ukkat B, Lorenz K, Hinze R, Thomusch O & Dralle H 2001 Importance of early screening and prophylactic thyroidectomy in asympomatic non index germline carriers. World Journal of Surgery 25 713-717.[Medline]

Vasen HF, Nieuwenhuijzen Kruseman AC, Berkel H, Beukers EK, Delprat CC, van Doorn RG, Geerdink RA, Haak HR, Hackeng WH, Koppeschaar HP et al. 1987 Multiple endocrine neoplasia syndrome type 2: the value of screening and central registration. A study of 15 kindreds in the Netherlands. American Journal of Medicine 83 847-852.[CrossRef][Web of Science][Medline]

Verdy M, Weber AM, Roy CC, Morin CL, Cadotte M & Brochu P 1982 Hirschprung's disease in a family with multiple endocrine neoplasia type 2. Journal of Pediatric Gastroenterology and Nutrition 1 603-607.[Web of Science][Medline]

Vestergaard P, Vestergaard EM, Brockstedt H & Christiansen P 2007 Codon Y791F mutations in a large kindred: is prophylactic thyroidectomy always indicated? World Journal of Surgery 31 996-1001.[Medline]

Wells SA Jr, Baylin SB, Leight GS, Dale JK, Dilley WG & Farndon JR 1982 The importance of early diagnosis in patients with hereditary medullary thyroid carcinoma. Annals of Surgery 195 595-599.[Web of Science][Medline]

Wells SA Jr, Chi DD, Toshima K, Dehner LP, Coffin CM, Dowton SB, Ivanovich JL, DeBenedetti MK, Dilley WG & Molley JF 1994 Predictive DNA testing and prophylactic thyroidectomy in patients at risk for MEN type 2A. Annals of Surgery 220 237-250.[Web of Science][Medline]

Williams EO 1963 A review of 17 cases of carcinoma of the thyroid and pheochromocytoma. Journal of Clinical Pathology 18 288-292.

Yip L, Cote GJ, Shapiro SE, Ayers GD, Herzog CE, Sellin RV, Sherman SI, Gagel RF, Lee JE & Evans DB 2003 Multiple endocrine neoplasia type 2: evaluation of the genotype-phenotype relationship. Archives of Surgery 138 409-416.[Abstract/Free Full Text]

You YN, Lakhani V & Wells SA 2007 New directions in the treatment of thyroid cancer. Journal of the American College of Surgeons 205 S45-S48.




This article has been cited by other articles:


Home page
JCOHome page
M. E. Robson, C. D. Storm, J. Weitzel, D. S. Wollins, and K. Offit
American Society of Clinical Oncology Policy Statement Update: Genetic and Genomic Testing for Cancer Susceptibility
J. Clin. Oncol., February 10, 2010; 28(5): 893 - 901.
[Full Text] [PDF]


This Article
Right arrow Abstract Freely available
Right arrow Full Text (PDF)
Right arrow Alert me when this article is cited
Right arrow Alert me if a correction is posted
Services
Right arrow Similar articles in this journal
Right arrow Similar articles in PubMed
Right arrow Alert me to new issues of the journal
Right arrow Download to citation manager
Citing Articles
Right arrow Citing Articles via HighWire
Right arrow Citing Articles via Web of Science (3)
Right arrow Citing Articles via Google Scholar
Google Scholar
Right arrow Articles by Sakorafas, G. H
Right arrow Articles by Peros, G.
Right arrow Search for Related Content
PubMed
Right arrow PubMed Citation
Right arrow Articles by Sakorafas, G. H
Right arrow Articles by Peros, G.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS