Skip to main content
Erschienen in: Endocrine Pathology 1/2021

Open Access 02.12.2020

Genomics and Epigenomics in Parathyroid Neoplasia: from Bench to Surgical Pathology Practice

verfasst von: C. Christofer Juhlin, Lori A. Erickson

Erschienen in: Endocrine Pathology | Ausgabe 1/2021

Abstract

The majority of parathyroid disease encountered in routine practice is due to single parathyroid adenoma, of which the majority arise as sporadic tumors. This is usually a straightforward diagnosis in endocrine pathology when in the appropriate clinical setting, although subsets of cases will exhibit atypical histological features that may warrant additional immunohistochemical and genetic analyses to estimate the malignant potential. Parathyroid carcinomas on the other hand, are bona fide malignant tumors characterized by their unequivocal invasion demonstrated through routine histology or metastasis. The ultimate endpoint for any molecular marker discovered through laboratory investigations is to be introduced in clinical routine practice and guide the surgical pathologist in terms of diagnostics and prognostication. For parathyroid tumors, the two main diagnostic challenges include the distinction between parathyroid adenoma and parathyroid carcinoma, as well as the pinpointing of hereditable disease for familial screening purposes. While numerous markers on genetic, epigenetic, and protein levels have been proposed as discriminative in these aspects, this review aims to condense the scientific coverage of these enigmatic topics and to propose a focused surgical pathology approach to the subject.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

General Background

In the clinical setting, the tumor responsible for primary hyperparathyroidism (PHPT) is usually a parathyroid adenoma. The vast majority of parathyroid adenomas are functioning due to an altered set point in terms of calcium sensing mechanisms, and the ensuing parathyroid hormone (PTH) secretion leads to hypercalcemia that may cause diverse symptoms in the afflicted patient [1]. However, the majority of parathyroid adenomas are identified through serum calcium screening. The peak incidence is among 50–60-year-old individuals, and the female to male ratio increases with increased age at diagnosis, reaching 5:1 among patients > 75 years of age [2]. The treatment is surgical, and cure rates at tertiary centers are usually high [3]. Most cases are preoperatively localized using combinations of various imaging techniques, such as neck ultrasound, single-photon emission computed tomography (SPECT/CT), and/or technetium (99mTc) sestamibi scintigraphy, and the endocrine surgeon can thus plan for a focused parathyroidectomy [4, 5]. Although the bulk of PHPT cases are sporadic tumors arising through the somatic acquisition of genetic aberrancies in driver genes, approximately 5% of cases are associated with familial disease. If a familial syndrome is suspected, a four-gland exploration with subtotal parathyroidectomy or total parathyroidectomy with intramuscular reimplantation is often the preferred strategy, as these patients may develop multiglandular, metachronous disease—and some also carry a risk of developing parathyroid carcinoma [6, 7].
From a histopathological perspective, parathyroid adenomas are usually well-circumscribed tumors composed of chief cells arranged in micro-acinar or palisading formations (Fig. 1a) [8]. Subsets of cells can exhibit hyperchromatic nuclei with nuclear atypia, and multinucleated tumor cells are sometimes observed. Mitotic figures can be seen in parathyroid adenoma and parathyroid carcinoma. The stromal fat content is reduced compared to normal gland histology, and a rim of normal appearing (although suppressed) parathyroid tissue can usually be seen, particularly in smaller parathyroid adenomas and less commonly seen in larger adenomas (Fig. 1a). What appears to be a rim of normal-appearing parathyroid tissue cannot be used to differentiate parathyroid adenoma from multiglandular disease, as up to 10% of parathyroid “hyperplasia” (multiglandular disease) may have what appear to be rims of normal parathyroid. Moreover, as intraoperative frozen section analyses not always are effective in distinguishing single parathyroid adenomas from multiglandular involvement, intraoperative PTH assays are probably more reliable in this context [9]. In order to diagnose a parathyroid adenoma, there must be no signs of malignant behavior, such as vascular or perineural invasion. Regarding their immunophenotype, most parathyroid adenomas are positive for chromogranin A, PTH, and GATA3. Numerous histological variants have been described, of which oncocytic parathyroid adenomas are the most common, followed by unusual variants such as parathyroid lipoadenomas and water-clear cell adenomas [8, 1012]. From a clinical perspective, oncocytic features associate to larger tumor size. Oncocytic parathyroid tumors are functional but may not have as elevated serum calcium or parathyroid hormone levels as comparable conventional parathyroid adenomas. Lipoadenomas might possibly correlate to the presence of arterial hypertension—suggesting that these subclassifications might disclose clinical considerations of importance [11, 13].
Subsets of parathyroid tumors exhibit atypical features usually observed in carcinomas, but yet lacking unequivocal criteria of malignancy such as invasive growth into periparathyroidal tissues (thyroid gland or soft tissues), lymphovascular or perineural invasion, regional and/or distant metastases. These features may include a trabecular growth pattern, fibrotic bands, tumor cells within the capsule, increased mitotic/proliferative activity, and nuclear atypia with macronucleoli (Fig. 1b, c) [8, 1416]. Parathyroid lesions with these features are termed “atypical parathyroid tumors” and could be considered tumors with unknown malignant potential—even though the majority of these cases will behave benign in a clinical context, with few recurrences following parathyroidectomy [17, 18]. Due to the fact that small subsets of atypical tumors will recur as metastatic parathyroid carcinomas, numerous studies have tried to highlight the malignant potential of this tumor subgroup using various combinations of histology and molecular markers, which are discussed in detail below.
Parathyroid carcinoma is the malignant form of PHPT. Quite rare, accounting for < 1% of PHPT, parathyroid carcinoma is an entity which from an endocrine pathology perspective always is feared, often discussed, but rarely diagnosed (personal observations) [8, 19]. Unlike parathyroid adenomas, which are more common in women than in men, parathyroid carcinomas affect woman and men equally. Clues to a parathyroid carcinoma diagnosis can be obtained preoperatively, as these tumors often are larger than adenomas, may be clinically palpable, and usually are associated with high serum calcium levels (often > 13.5 mg/dl) with patients exhibiting various symptoms related to hypercalcemia, such as nephrolithiasis and bone disease [20, 21]. Perioperative findings of the tumor being adherent to adjacent structures can also be information of value when assessing these rare lesions, and for this reason, surgeons usually opt in for an en bloc resection of the afflicted ipsilateral thyroid lobe when suspecting parathyroid carcinoma (Fig. 1d) [21]. However, one must be cautious in evaluating a parathyroid that may be adhesed to adjacent structures such as the thyroid gland, as parathyroid adenomas may also be firmly adhesed to this organ. The difference of course is that parathyroid adenomas are not invasive of the thyroid while parathyroid carcinomas may be invasive of the adjacent thyroid. Parathyroid carcinomas exhibit local invasive behavior and may spread locally to adjacent structures and later on to distant sites (Fig. 1e, f) [8]. Chemotherapy and/or radiotherapy are largely ineffective, and the 10-year survival rate is around 50–70% [22], with death often due to hypercalcemia. Morbidity due to complications following repetitive neck surgery (hypoparathyroidism, recurrent nerve palsy) is also high [23].
The differentiation of parathyroid carcinoma from other carcinomas in the neck is usually straightforward. Parathyroid carcinomas are neuroendocrine tumors and generally positive for chromogranin A. Other neuroendocrine tumors in the neck such as medullary thyroid carcinoma will also shows staining for chromogranin A, but medullary thyroid carcinomas are usually positive for thyroid transcription factor 1 while negative for parathyroid hormone. Calcitonin is usually positive in medullary thyroid carcinoma and usually negative in parathyroid carcinoma; however, unusual staining patterns can be seen such an exceptional case of parathyroid carcinoma positive for calcitonin and calcitonin gene related peptide [24]. Moreover, practicing pathologist should also be aware that medullary thyroid carcinomas can express PAX8 in a clone-dependent manner, in which absence of immunoreactivity is noted when monoclonal PAX8 antibodies are applied, as opposed to positive staining using polyclonal antibodies [25, 26]. As parathyroid tissue might stain positive for polyclonal PAX8 antibodies, we recommend a distinguishing panel of PTH and TTF1 when assessing these differentials [27].
Although a plethora of epigenetic and genetic aberrancies have been identified in parathyroid tumors, few markers have paved their way into clinical routine practice. In this review, we focus on two main clinical predicaments concerning parathyroid tumors, namely, (1) the identification of tumors associated to various underlying syndromes of importance for genetic counseling, and (2) the distinction between benign and malignant tumors to triage each patient to correct follow-up and treatment algorithms. In the following sections, we highlight molecular markers of importance that facilitate these diagnostic quandaries, and discuss their potential as discriminative clinical markers of value to the surgical pathologist.

Familial PHPT: Underlying Causes

Much of what we today know about mutational driver events in parathyroid tumorigenesis stems from earlier work in kindreds with familial PHPT, long before the appearance of next-generation sequencing techniques. By genetic linkage analyses of family members with autosomal dominant PHTP, candidate gene loci segregating with disease-afflicted individuals were identified, followed by identification of mutational events by cumbersome Sanger sequencing of a large number of candidate genes within these regions. By this methodology, germline mutations of the MEN1 (multiple endocrine neoplasia type 1), RET (rearranged during transfection), and CDC73 (cell division cycle protein 73) (originally entitled hyperparathyroidism type 2, HRPT2) genes as events underlying the development of multiple endocrine neoplasia type 1 (MEN1), multiple endocrine neoplasia type 2A (MEN2A), and hyperparathyroidism-jaw tumor (HPT-JT) syndromes, respectively (Table 1) [2836]. These three conditions exhibit high prevalence of PHPT, occurring in approximately 90% of MEN1 kindred, 20–30% of MEN2A kindred, and in 80% of HTP-JT kindred [8]. In MEN1 patients, PHPT is the most common disease manifestation, followed by pituitary tumors (30–40% of patients), duodenal and pancreatic neuroendocrine tumors (most often gastrinoma, insulinoma and/or glucagonoma, 40% of patients), and adrenocortical lesions (20–45% of patients) [8, 37]. In MEN2A, most patients (> 90%) develop medullary thyroid carcinoma and pheochromocytoma (50%), whereas PHPT is more uncommon, occurring in 15–30% of patients [8, 38]. For HPT-JT kindred, 70–80% develop PHPT, whereas approximately 10% of patients also develop ossifying fibromas of the mandible or maxilla [8, 39]. PHPT in MEN1 usually presents as multiglandular disease that may develop in synchronous or metachronous settings and may be asymmetric. For the MEN2A syndrome, the PHPT may present as multiglandular or single gland disease. There are exceedingly few reports of unequivocal parathyroid carcinomas arising in MEN1 or MEN2A kindred [4042]. The hyperparathyroidism in HPT-JT syndrome is usually associated with a parathyroid adenoma; however, 15–40% of patients carrying CDC73 mutations or gene deletions will develop parathyroid carcinoma [8, 43, 44].
Table 1
Overview of general constitutional and somatic genetic aberrations in parathyroid adenomas and carcinomas
 
MEN1 mut.
RET mut.
CDC73 mut.
CaSR mut.
GCM2 mut.
CDKi family mut.
PTH-CCND1 inversion
CTNNB1 mut.
EZH2 mut.
mtDNA mut.
TP53 mut.
TERTp mut.
RIZ1 meth.
RASSF1A meth.
APC meth.
Familial forms
  MEN1
1
-
-
-
-
-
-
-
-
-
-
-
-
-
-
  MEN2a
-
1
-
-
-
-
-
-
-
-
-
-
-
-
-
  HPT-JT
-
-
1
-
-
-
-
-
-
-
-
-
-
-
-
  FHH1
-
-
-
1
-
-
-
-
-
-
-
-
-
-
-
  FIHP
2
-
3
3
3
-
-
-
-
-
-
-
-
-
-
  MEN1 variants
-
-
-
-
-
1
-
-
-
-
-
-
-
-
-
Sporadic PHPT
  PA
3
-
4
-
-
4
4
4
4
2
-
-
3
1
2
  PC
4
-
2
-
-
-
-
-
-
-
4
4
-
-
2
mut mutation, meth aberrant promoter methylation, CDKi cyclin-dependent kinase inhibitor, mtDNA mitochondrial DNA, TERTp TERT promoter, PA parathyroid adenoma, PC parathyroid carcinoma
Germline
Somatic
(1) Almost always present (81–100%)
(2) frequently present (41–80%)
(3) sometimes present (11–40%)
(4) rarely present (≤ 10%)
-; not reported
Apart from these three syndromes, hyperparathyroidism is also seen as the sole feature in familial isolated hyperparathyroidism (FIHP). These families often, but not always, present with germline mutations in either MEN1, CDC73, or the calcium sensing receptor (CaSR) gene, of which the latter is also associated to the development of familial hypocalciuric hypercalcemia type 1 (FHH1) [4550]. The reason why some patients with germline mutations develop full-blown MEN1, HPT-JT, and FHH1 syndromes, while others develop FIHP, is not clearly understood—as there is no apparent genotype to phenotype correlation in terms of mutation types and exon localization. Recently, germline activating glial cells missing transcription factor 2 (GCM2) gene mutations were also coupled to FIHP, adding yet another candidate to the growing palette of genes underlying the development of familial hyperparathyroidism [51]. Moreover, germline inactivating mutations in cyclin-dependent kinase inhibitor (CDKI) genes have been found in rare families with MEN1-like syndromes (with mutations in either CDKN1A, CDKN2B, or CDKN2C) or the MEN4 syndrome, a phenotypic MEN1 syndrome characterized by mutations in CDKN1B [50, 52, 53].

Somatic Genetics in Parathyroid Adenomas

Given the identification of the MEN1, RET, and CDC73 gene aberrancies as main responsible for the development of familial PHPT, numerous studies followed in which the involvement of these genes were assessed in sporadic parathyroid tumors. Approximately 25–40% of all sporadic parathyroid adenomas harbor LOH of the MEN1 gene locus at 11q13, and half of these cases also exhibit an inactivating MEN1 mutation of the remaining allele (Table 1, Fig. 2) [54, 55]. Interestingly, an association between somatic MEN1 mutations and mild PHPT symptoms has been observed, possibly arguing for early-stage events in parathyroid tumorigenesis [55]. Moreover, while somatic CDC73 gene mutations have been reported in small subsets of sporadic parathyroid adenomas, no reports on somatic RET gene mutations in parathyroid adenoma have been noted [35, 56, 57].
Moreover, mutational and/or epigenetic silencing of other genes predisposing for FIHP and MEN1-like syndromes have also been detected in small subsets of apparent sporadic parathyroid adenomas, including CDKN1A, CDKN1B, CDKN2A, CDKN2B, CDKN2C, and GCM2 [53, 5860]. Of particular interest, CDKN1B encodes p27 and down-regulation of p27 has been described in PAs, both on the RNA- and protein level [6163]. Moreover, CDKN1B mutations have also been functionally linked to the development of parathyroid tumors, thereby solidifying the role of aberrant cell cycle regulation in the development of parathyroid adenomas [64]. Continuing on the cell cycle topic, a recurrent chromosomal inversion involving the peri-centromeric portion of chromosome 11 has been observed in exceedingly small subsets of sporadic parathyroid adenomas. This rearrangement causes the juxtaposition of the PTH 5′ regulatory region to the CCND1 oncogene coding region, causing a constitutively expression of the CCND1 corresponding protein cyclin D1. Although rare in sporadic parathyroid adenoma, overexpression of cyclin D1 is a common event, and therefore, other mechanisms apart from rare chromosomal inversions involving CCND1 or mutations in cyclin D1-regulating CDKIs are expected to play a role. Instead, promoter hypermethylation and down-regulation of various CDKIs could probably explain the commonly observed cyclin D1 upregulation in parathyroid adenomas (Fig. 2) [65].
Apart from the discovery of somatic alterations in established parathyroid adenoma susceptibility genes discussed above, the advent of next-generation sequencing techniques has led to the discovery of additional gene mutations of possible importance to parathyroid adenoma development. By whole-exome sequencing of parathyroid adenomas, an activating missense mutation in the methyltransferase gene enhancer of zeste homolog 2 (EZH2) was detected in one out of 8 adenomas interrogated, and additional targeted sequencing of 185 adenomas revealed one additional case with the same mutation [66]. The EZH2 gene is an epigenetic regulator of chromatin accessibility with an association to tumorigenesis in general, thereby providing additional validity of this gene constituting a possible contributor of parathyroid adenoma development—which was also verified using functional experiments in a parathyroid cell line [67, 68]. Since then, additional whole-exome sequencing studies have corroborated low frequencies of EZH2 mutations in sporadic adenomas [69].
Additional mutational events occurring in low frequencies of parathyroid adenomas include activating CTNNB1 mutations, of which some have been reported as homozygous in small numbers [70], although not reproduced by others [7173]. CTNNB1 encodes beta-catenin, a central onco-protein regulating the Wingless type (Wnt) pathway, and data suggests that nuclear accumulation of beta-catenin could be an important player in the development of parathyroid adenomas—either through activating mutations or through aberrantly expressed Wnt co-receptors (Fig. 2) [74].
On the epigenetic level, apart from hypermethylation of CDKIs mentioned above, aberrant methylation has also been reported for numerous tumor-related genes, including WT1, SFRP1, SFRP2, SFRP4, RIZ1, APC, and RASSF1A [59, 65, 7577]. Most notably, RASSF1A hypermethylation was strongly associated to down-regulation on the mRNA level in virtually all parathyroid adenomas, thereby constituting one of the most commonly known genetic aberrancies in this disease (Table 1) [75]. Moreover, on a global level, adenomas seem to exhibit similar levels of methylation as non-tumorous parathyroid tissues, suggesting that epigenetic dysregulations are driven by gene-specific events and not due to a general hypo- or hypermethylation pattern [75].
Regarding specific parathyroid adenoma subtypes, there is also an established correlation between oncocytic parathyroid adenomas and somatic mutations in genes encoded by mitochondrial DNA (mtDNA), especially mitochondrial respiratory chain complex genes NADH dehydrogenase 1, 4, and 5 (ND1, ND4, and ND5) (Table 1, Fig. 2). As oncocytic tumors in general exhibit prominent amounts of mitochondria, the association is intriguing [78]. However, as no recurrent mutations were observed, these findings mandate functional verification before a true driver status could be assigned to any of these alterations.

Somatic Genetics in Parathyroid Carcinomas

Somatic CDC73 gene mutations are the most frequent somatic alteration in parathyroid carcinoma (Table 1, Fig. 2) [7982]. These mutations are in general disruptive due to premature truncations or frameshift alterations, alternatively the mutations are of missense nature in conserved regions encoding the nuclear localization signals (NLSs) or the human polymerase–associated factor 1 (hPAF1) complex of the corresponding protein product, termed parafibromin. Apart from mutations, LOH encompassing the CDC73 gene locus and aberrant CDC73 promoter methylation have also been reported as somatic events in parathyroid carcinoma [8385].
Parafibromin is a member of the hPAF regulatory complex, a key transcriptional unit that interacts with RNA polymerase II and facilitates transcriptional activity due to histone-modifying and chromatin remodeling processes [86]. Parafibromin is associated with tumor-suppressive properties, as (a) the majority of CDC73 germline mutations in HPT-JT and FIHP kindred as well as the bulk of somatic mutations in sporadic parathyroid carcinomas are disruptive [35, 43, 7981, 8789], (b) the majority of tumors with CDC73 mutations exhibit loss of parafibromin expression [57, 8995], and (c) functional experiments with CDC73 plasmids support an anti-proliferative effect of the wild-type protein [96]. Indeed, parafibromin has been found to regulate cyclin D1 levels, exhibit pro-apoptotic effects, regulate the Wnt pathway through interactions with beta-catenin, as well as the c-Myc oncogene through direct binding to the promoter region of this gene [96100]. Intriguingly, parafibromin also seems to exhibit oncogenic features in the presence of certain molecular partners, suggesting a yin-yang modus operandi of this protein [101].
In contrast to parathyroid adenomas, somatic MEN1 mutations are very infrequent in parathyroid carcinomas, but nonetheless reported [102]. Given the exceedingly low rate of malignant PHPT in MEN1 kindred, other genetic events apart from this aberrancy are expected to drive the invasive behavior in parathyroid carcinoma.
TP53 gene mutations are among the most common genetic aberrations in malignant epithelial tumors, although these genetic events seem to be unusual in parathyroid carcinomas [103105]. Even so, LOH of one TP53 allele seems to be more common [103]. Similarly, loss of the retinoblastoma 1 (RB1) gene, encoding pRB, is observed in the majority of parathyroid carcinomas, however inactivating mutations have not been reported [106108]. The P53 and pRB proteins are regulators of cell cycle progression and two bona fide tumor suppressors usually required to be silenced on both alleles in order to promote neoplasia, and given the fact that parathyroid carcinomas recurrently exhibit absent pRB expression, additional inactivating mechanisms apart from mutations are most likely operational [106].
Next-generation sequencing studies on parathyroid carcinomas are rare, not surprising given the low prevalence of this disease in general. In a recently published whole-genome sequencing study of 23 parathyroid carcinomas, the authors conclude that CDC73 gene mutations were the most common sequence aberration, occurring in almost 40% of cases. Increased copy number variants were seen in parathyroid carcinomas with CDC73 mutations, and these cases also carried an increased tumor mutational burden and poorer patient outcome. In unrelated, exome-based studies, recurrent mutations in AarF domain containing kinase 1 (ADCK1) and prune homolog 2 with BCH domain (PRUNE2) have been reported in parathyroid carcinomas; however, their functional roles has not been elucidated [109, 110]. Moreover, a general overrepresentation of mutations in genes associated with DNA repair and cell cycle regulation seems evident [111, 112], and rare mutations in established cancer-associated genes have also been identified, for example succinate dehydrogenase complex flavoprotein subunit A (SDHA) and DICER1. From a therapeutic perspective, the majority of parathyroid carcinomas might carry alterations suitable for molecular targeted therapies, thus highlighting the potential role for next-generation sequencing as a tool to identify cases with potential for targeted therapeutic interventions [113].
Promoter mutations of the telomerase reverse transcriptase (TERT) gene are heavily implicated in human cancers, as they convey increased TERT expression, which in turn promotes immortalization. However, these mutations seem fairly rare in parathyroid carcinomas, although these tumors in general express TERT protein [112, 114, 115]. As of this, alterative mechanisms leading to increased TERT expression in parathyroid carcinomas are suspected.
Aberrant epigenetic mechanisms are also at play in parathyroid carcinomas. Via global methylome analyses, parathyroid carcinomas and adenomas seem to exhibit hypermethylation of CDKN2B, CDKN2A, WT1, SFRP1, SFRP2, and SFRP4 [59]. Moreover, beside the recurrent CDC73 promoter hypermethylation discussed above, altered methylation levels of the adenomatous polyposis coli (APC) promoter 1A region is recurrently seen in parathyroid tumors, although the APC mRNA expression seem to be retained by an unmethylated 1B promoter region [75, 116]. In parathyroid carcinomas specifically, loss of APC protein expression is a frequent event, and this could in part be due to aberrant methylation patterns rather than gene mutations [116118]. The APC protein is a tumor suppressor that regulates the Wnt pathway, and loss of APC expression is therefore thought to stimulate proliferation in parathyroid cells. In more recent years, the regulation of epigenetic de-methylation has been highlighted in cancer, especially the discovery of the TET1/TET2 enzymatic activity catalyzing oxidation of 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC). In parathyroid carcinomas, 5hmC levels and TET1 expression have been found extensively reduced, suggesting a general reduction in de-methylation events across the genome [119, 120]. As this phenomenon is tightly linked to the presence of TERT promoter mutations in unrelated cancer types, the general absence of these mutations in parathyroid carcinoma would suggest that other molecular mechanisms influence the observed lack of global de-methylation—a subject worthy of follow-up studies [121].

The Diagnostic Challenges

The MEN1 syndrome is by far the most common among the hereditary conditions detailed in this review, with one case per 40,000 as opposed to MEN2A (1 case per 2 million) and the HPT-JT syndrome (unknown prevalence, but expected to be exceedingly low) [8]. Therefore, surgical pathologists will most likely diagnose MEN1 related parathyroid adenomas to a much larger extent than MEN2A, HPT-JT, FIHP, or MEN1-like related cases. Although the MEN1 syndrome often is diagnosed long before the patient is subjected to parathyroidectomy, the non-total penetrance in younger years as well as the occurrence of de novo MEN1 mutations in subsets of patients with healthy parents allow for subsets of patients being misclassified as sporadic PHPT patients. As up to 10% of individuals with primary parathyroid “hyperplasia” (multiglandular disease, adenomatosis, multiple adenomas) have MEN1, genetic screening for familial disease has been considered for all patients with primary parathyroid “hyperplasia.” From a diagnostic pathology standpoint, is there a way for the attentive pathologist to aid in the detection of syndromic PHPT in cases where the syndromic association was not evident preoperatively?
MEN1 was early on assigned a tumor-suppressor gene status, as the bulk of mutational events in MEN1 kindred reported represent inactivating events leading to premature truncations of the menin protein [122]. Moreover, most parathyroid adenomas arising in MEN1 patients harbor loss of heterozygosity (LOH) of the MEN1 wild-type allele on the somatic level, thereby arguing in favor for the Knudson’s “two hit” theory in which bi-allelic inactivation of a tumor suppressor gene is needed in order for a tumor to develop [123125]. Therefore, the question arises if clinical screening using expressional analyses targeting menin could be a cheap and efficient way for the surgical pathologist to triage menin-negative PHPT cases for genetic screening—as loss of menin expression would be indicative of MEN1 gene aberrancies.
Menin is a predominantly nuclear protein with a highly conserved sequence among species, and the protein is universally expressed in most human tissues [122]. The protein harbors nuclear localization signals and leucine zipper motifs, both of which are features needed to regulate gene expression though direct interaction with DNA elements. For example, menin has been shown to interact with JunD, a proto-oncogene encoding a transcription factor regulating apoptosis and TP53 gene activation (Fig. 2) [126]. Moreover, menin has been proposed as an important player in the regulation of various signaling networks associated to tumor development, such as the TGF-beta, RAS, Wingless type (Wnt) pathways—as well as influencing the expression of the telomerase reverse transcriptase (TERT) gene [127130].
From a histological context, the MEN1-related parathyroid disease is often multiglandular, although not always synchronous in presentation [8]. Unlike sporadic parathyroid adenomas, MEN1-related tumors are often devoid of a rim of normal parathyroid tissue, and they may be composed of chief cells arranged in compact, sheet-like formations [8, 131]. The tumors may exhibit fibrosis and have similar appearance as parathyroid hyperplastic lesions arising in the setting of chronic renal failure, and almost always lack invasive features. Overall, findings of multiglandular disease and absence of a normal parathyroid rim in a PHPT patient should raise the suspicion of the MEN1 syndrome; however, a “rim” can also be seen in up to 10% of multiglandular disease. In all, there is no histological feature that sets MEN1-related adenomas apart from sporadic ones.
Expression studies regarding menin and associations to underlying MEN1 mutational status in parathyroid tumors have been promising, as menin immunohistochemistry seem to exhibit high sensitivity to detect underlying MEN1 mutations and/or gene deletions [132, 133]. Even so, the interpretation of menin immunohistochemistry is not straightforward, and the lack of comprehensive studies on the subject makes menin immunohistochemistry inappropriate for clinical routine screening purposes. Moreover, as the MEN1 gene is frequently deleted and mutated also on the somatic level in sporadic parathyroid adenomas, additional factors are needed to bring into consideration before suspecting MEN1 syndrome–related PHPT from expressional analyses in the pathology laboratory. For example, the penetrance of PHPT in MEN1 patients reaches 95% at 50 years of age, and therefore, the diagnosis of MEN1 in older patients with multiglandular disease should not be completely overlooked—although the bar for considering an MEN1 diagnosis should be even lower in adolescent patients [37]. In all, a concerted teamwork between endocrine surgeons and pathologists covering the patient’s medical history, disease presentation, radiology, and histopathology is probably needed to properly identify all MEN1-related PHPT in the clinical setting.

HPT-JT-Associated PHPT: Clues from the Histopathological Workup?

As HPT-JT patients carry germline CDC73 gene mutations or deletions, it comes as no surprise that the prevalence of parathyroid carcinoma in this patient category is much higher (15–30%) than in unselected PHPT patient cohorts (< 1%). As parathyroid carcinoma is so uncommon, it has been suggested that individuals diagnosed with parathyroid carcinoma should be promptly evaluated for HPT-JT. Additionally, if the pathologist raises the suspicion that the patient is indeed an HPT-JT kindred, a raised awareness of the increased parathyroid carcinoma risk is mandated when diagnosing the tumor. Easily recognizable features such as a tumor being cystic may in itself raise the possibility of HPT-JT syndrome. In the most comprehensive study yet regarding morphological features of CDC73 mutated parathyroid tumors, the authors conclude that these lesions are characterized by sheet-like, compact growth rather than the usual acinar patterns visualized in the bulk of parathyroid tumors, as well as a typical eosinophilic cytoplasm distinct from the granular oxyphilic cell type often observed in areas of parathyroid adenomas [89]. Many tumors also exhibited enlarged nuclei and a perinuclear cytoplasmic clearing. In all, these findings suggest that the occurrence of these histological parameters might suggest the occurrence of an underlying CDC73 gene mutation, and these cases should preferably be investigated more thoroughly with parafibromin immunohistochemistry and/or CDC73 gene sequencing.

Pinpointing Parathyroid Carcinoma in the Diagnostic Setting

Parathyroid carcinoma is the malignant form of PHPT. It constitutes only 0.5–3% of PHPT cases. As many parathyroid tumors present with various degrees of atypia, although not fulfilling the current WHO criteria for parathyroid carcinoma, there is a great need to identify malignant potential in these lesions by other means than histology alone. Moreover, any clinical marker of value in this context would need to exhibit very high specificity, in order to avoid falsely diagnosing benign tumors as carcinoma. The bulk of studies on this subject have attempted to address this dilemma via immunohistochemistry, which is a gold standard, inexpensive methodology used in most pathology laboratories and therefore suitable for rapid implementation in routine clinical practice [27].
One of the earliest immunohistochemical markers proposed to identify parathyroid carcinoma was Ki-67, an extensively used proliferation marker used to identify cells in active (non-G0) phases of the cell cycle. Although there seem to be an overrepresentation of carcinomas among high-proliferative parathyroid tumors, the overlap between parathyroid adenomas and carcinomas is considerable [134139]. In a similar fashion, investigations of the cell cycle protein cyclin D1 was initially proven of value, as it was found upregulated in most parathyroid carcinoma compared to adenomas [140]. However, subsequent studies have identified overlap in expression between benign and malignant groups [117, 141].
Detection of widespread LOH of the RB1 locus in parathyroid carcinoma with loss of its corresponding protein pRB prompted the investigation of pRB expression in parathyroid tumors—but with divergent results both concerning staining outcomes as well as LOH events [108, 134, 135, 142, 143]. In short, while LOH of the RB1 locus seem to confer high sensitivity for the detection of parathyroid carcinoma, a significant amount of adenomas harbor the same genetic aberrancy, and the reduced specificity renders this marker of less value when screening for rare carcinomas in PHPT. Moreover, a positive pRb staining seems to be variable in terms of the proportion tumor cells stained, making the interpretation somewhat challenging [108].
Following the advent of tissue microarrays, a study concluded that the combined positivity of p27, B-cell lymphoma 2 (bcl-2), and mouse double minute 2 homolog (mdm2) adjoined by a low Ki-67 proliferation index indicated a benign clinical behavior of any given parathyroid tumor, as this profile was not evident in any parathyroid carcinoma but found in the vast majority of adenomas [136]. Both bcl-2 and mdm2 are members of the P53 pathway, thereby furthermore solidifying the relationship between aberrant P53 signaling and parathyroid carcinoma. However, follow-up studies have demonstrated overlaps between parathyroid adenomas and carcinomas, not least the finding of down-regulation of p27 in large subsets of parathyroid adenomas [63, 144146]. P53 immunoreactivity shows variable staining in benign and malignant parathyroid tumors and does not appear to be useful as a single marker in separating these tumors [144, 144, 147]. Other diagnostic nomograms have been evaluated to differentiate malignant from benign parathyroid neoplasms, including among others protein gene product 9.5 (PGP 9.5), Ki-67, galectin-3, E-cadherin, and pRb markers [148]. More novel combinations of immunohistochemical and in situ hybridization approaches have also been evaluated, such as long noncoding RNA expression [149].
The most well-studied and reproduced marker in the context of distinguishing parathyroid carcinoma from adenoma is parafibromin, the 531 amino acid protein product of the CDC73 main transcript [35]. Given the increased parathyroid carcinoma risk in HPT-JT kindred as well as the association between somatic CDC73 mutations and sporadic parathyroid carcinoma, subsequent immunohistochemical analyses regarding parafibromin early-on proved of value to detect the majority of CDC73 mutated cases, including most parathyroid carcinomas [90, 91, 9395, 118]. Moreover, the vast majority of parathyroid adenomas analyzed have been shown to retain parafibromin immunoreactivity, apart from subsets of sporadic cases with a predominant cystic growth pattern, HPT-JT-associated parathyroid adenomas, and atypical parathyroid tumors [17, 57, 93, 150]. Thus, with the exceptions of the previously mentioned exceptions, parafibromin is useful for screening purposes in the PHPT population and can be a highly useful marker of malignant potential in many cases. The value of this marker in the clinical setting has been assessed both by comprehensive meta-analyses and by recent reports from high-volume centers [89, 92, 151, 151].
As parafibromin is a predominant nuclear protein, loss of nuclear immunoreactivity is considered pathognomonic for an underlying inactivating CDC73 mutation. As most CDC73 mutations, be it germline or somatic, are either deleterious, located in early exons (causing premature termination) or missense alterations in important regulatory regions (such as the nuclear localization regions), mutated parafibromin is usually unable to reach its nuclear localization [97, 151, 152]. Most studies therefore seem to agree that almost all parathyroid tumors with wild-type CDC73 sequence display a diffuse positive parafibromin stain (Fig. 3a), while reduced or absent nuclear parafibromin expression is an aberrant staining pattern and might signal the presence of a mutation (Fig. 3b, d, e). While diffuse loss of parafibromin expression is usually seen, reduced (but not complete) nuclear parafibromin expression can also be noted in CDC73 mutated cases, occurring in subsets of tumor nuclei in a chessboard type of pattern (Fig. 3b) [92, 94]. Additionally, rare cases with parafibromin-negative nucleolar compartments have also been reported, which could be of importance given the nucleolar roles of wild-type parafibromin [88, 153157]. Specifically, subsets of parathyroid tumors with CDC73 mutations thought to disrupt the nucleolar localization signals might exhibit retained nuclear parafibromin immunoreactivity while evidently displaying negative nucleolar staining—and practicing pathologist should therefore be aware of this staining pattern as well (Fig. 3c) [156]. As most studies focus on the 2H1 monoclonal parafibromin antibody targeting the N-terminal, the usage of different antibodies should not explain these different staining patterns. Indeed, the “partial loss” pattern described above has been reported in cases assessed with four different parafibromin antibodies, suggesting that these observations are not biased by the selection of a specific target epitope [94]. The interpretation of parafibromin immunohistochemistry is therefore not completely straightforward, and additionally, the laboratory processing (including different antigen retrieval techniques and primary antibody incubation time) might affect the overall staining outcome [94, 131]. Moreover, parafibromin expression is often found stronger in the tumor periphery than in central aspects of the lesion, and therefore internal controls (such as endothelial cells) should be assessed and noted as positive before a negative parafibromin stain is called out (Fig. 3d, e) [93, 131].
Most parathyroid tumors will be assessed for parafibromin immunoreactivity if they are atypical, i.e., lack undisputable evidence of malignancy but display worrisome features (be it clinical or histological) often seen in parathyroid carcinomas. The preponderance of these tumors stain positive, although large subsets can be parafibromin deficient [17, 91, 94, 150]. How should these cases be interpreted, and what should be reported to the surgeon? Consulting the literature, it seems evident the vast majority parafibromin-negative atypical tumors will behave benign, even after long-term follow-up [17, 91]. Even so, the recurrence rate is not entirely negligible, and the finding of an aberrant parafibromin stain should also highlight the need to exclude an underlying germline CDC73 gene mutation—especially if there is a positive family history or if the proband itself exhibits multiglandular disease (Fig. 4) [17, 89, 91].
Given the non-perfect parafibromin sensitivity and specificity, researchers have been looking for additional markers to complement parafibromin immunohistochemistry. Shortly after parafibromin was coupled to the Wnt pathway, investigations of Wnt regulators detected widespread loss of APC immunoreactivity in most parathyroid carcinomas—which has later been coupled to promoter hypermethylation in PCs (Fig. 3f) [116, 117, 150]. As parathyroid adenomas are APC positive in general, the marker has gained ground as a clinical adjunct to parafibromin [117, 118]. However, as panels with two negative markers have its limitations in terms of interpretation associated to tissue fixation, researchers have looked for markers upregulated in parathyroid carcinomas. In this aspect, galectin-3 and PGP9.5 have both been shown to stain positive in the majority of parathyroid carcinomas while only expressed in few adenomas, and different panel combinations using galectin-3 and/or PGP9.5 with parafibromin have proven more reliable than using parafibromin alone [158-161].
Overall, the parathyroid carcinoma diagnosis is reserved for cases exhibiting unequivocal histological signs of invasive behavior, and no single molecular marker has yet been able to safely predict the malignant potential in atypical cases—which is most likely influenced by the general abundance of atypia in benign tumors and the rarity of truly metastatic cases. Moreover, there is also a potential sampling bias, as malignant tumors are not primarily resected if advanced disease is present at the time of diagnosis, and the fact that tumors with a molecular potential for spread might be resected well before their dissemination.

Discussion

The molecular background of parathyroid neoplasia is well characterized from a driver gene perspective, not least due to the identification of mutations in genes responsible for the development of hereditary PHPT. These genes were subsequently found aberrant also in sporadic cases, and still—some 20 years after original identification—enjoy their position as the top recurrently mutated genes in parathyroid tumors. Modern molecular analyses have since then expanded our knowledge regarding common dysregulations in parathyroid tumors, and highlighted both genetic and epigenetic changes involved in the development of PHPT. Next-generation sequencing analyses of both parathyroid adenoma and carcinoma cohorts have helped identify additional genes of potential impact for both groups, although it is clearly demonstrated that the “low-hanging fruit” in terms of highly recurrent events already have been picked.
From a clinical perspective, CDC73 mutations and loss of parafibromin expression have been firmly established as events coupled to parathyroid malignancy. Additionally, comprehensive sequencing of parathyroid carcinoma has identified mutations suitable for molecular targeted therapy as well as overall mutational burden as a prognostic tool of potential value, thereby highlighting the usage of modern genetic analyses where histology and immunohistochemistry might be insufficient. Given the rapid evolution of molecular techniques used for complimentary analyses in the routine clinical setting, the combination of histology, immunohistochemistry and next-generation sequencing might constitute standard work-up for parathyroid tumors displaying significant atypia in the near future.
To summarize the field for the surgical pathologist, parathyroid adenoma is usually a straightforward diagnosis, but there are histological patterns coupled to underlying syndromes worth remembering—as they could help identify hereditary disease of importance for clinical follow-up. When present, histologically atypical features in a parathyroid lesion could signify malignant potential, but invasive growth is still the only accepted criteria to diagnose parathyroid carcinoma. A plethora of immunohistochemical markers could potentially aid in the identification of parathyroid carcinoma, but many display subpar specificity and should be interpreted with caution. Loss of parafibromin nuclear expression correlates with the presence of an underlying CDC73 gene mutation, which in turn is coupled to an increased risk of developing parathyroid carcinoma. The mutation might also be present in germline tissues and thus predispose to familial disease.
Even though much of the genetic landscape of parathyroid tumors has been studied, two key queries remain to be deciphered, namely (1) the identification of additional driver genes responsible for the development of parathyroid adenomas, and (2) the quest for additional molecular events driving the malignant potential of parathyroid tumors besides parafibromin. As approximately half of parathyroid adenomas lack mutations in the most common driver genes, additional genetic events are expected to be found. Moreover, as the majority of HPT-JT kindred develop adenomas, other genetic aberrations apart from the inactivation of CDC73 are likely to be required to propel the invasive behavior in parathyroid carcinomas. Hopefully, these questions can be solved in the future by comprehensive, next-generation sequencing studies using multi-center tumor cohorts.

Acknowledgements

The authors are thankful to Dr. Anders Höög, Karolinska Institutet, Stockholm, Sweden, for providing macroscopic and microscopic images of parathyroid tumor specimen.

Compliance with Ethical Standards

Conflict of Interest

The authors declare that they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
3.
Zurück zum Zitat Donatini G, Marciniak C, Lenne X, Clément G, Bruandet A, Sebag F et al, on the behalf of AFCE Study Group (2020) Risk Factors of Redo Surgery After Unilateral Focused Parathyroidectomy: Conclusions From a Comprehensive Nationwide Database of 13,247 Interventions Over 6 Years. Ann Surg. 272:801-806. https://doi.org/10.1097/SLA.0000000000004269 Donatini G, Marciniak C, Lenne X, Clément G, Bruandet A, Sebag F et al, on the behalf of AFCE Study Group (2020) Risk Factors of Redo Surgery After Unilateral Focused Parathyroidectomy: Conclusions From a Comprehensive Nationwide Database of 13,247 Interventions Over 6 Years. Ann Surg. 272:801-806. https://​doi.​org/​10.​1097/​SLA.​0000000000004269​
4.
Zurück zum Zitat Patel SG, Saunders ND, Jamshed S, Weber CJ, Sharma J (2019) Multimodal Preoperative Localization Improves Outcomes in Reoperative Parathyroidectomy: A 25-Year Surgical Experience. Am Surg 85:939–943. https://doi.org/10.1177/000313481908500932CrossRef Patel SG, Saunders ND, Jamshed S, Weber CJ, Sharma J (2019) Multimodal Preoperative Localization Improves Outcomes in Reoperative Parathyroidectomy: A 25-Year Surgical Experience. Am Surg 85:939–943. https://​doi.​org/​10.​1177/​0003134819085009​32CrossRef
6.
Zurück zum Zitat Nastos C, Papaconstantinou D, Kofopoulos-Lymperis E, Peppa M, Pikoulis A, Lykoudis P et al (2020) Optimal extent of initial parathyroid resection in patients with multiple endocrine neoplasia syndrome type 1: A meta-analysis. Surgery. S0039-6060(20)30553-5. https://doi.org/10.1016/j.surg.2020.08.021 Nastos C, Papaconstantinou D, Kofopoulos-Lymperis E, Peppa M, Pikoulis A, Lykoudis P et al (2020) Optimal extent of initial parathyroid resection in patients with multiple endocrine neoplasia syndrome type 1: A meta-analysis. Surgery. S0039-6060(20)30553-5. https://​doi.​org/​10.​1016/​j.​surg.​2020.​08.​021
8.
Zurück zum Zitat Lloyd RV, Osamura RY, Klöppel G, Rosai J (2017) International Agency for Research on Cancer WHO classification of tumours of endocrine organs, 4th edition. International Agency for Research on Cancer, Lyon Lloyd RV, Osamura RY, Klöppel G, Rosai J (2017) International Agency for Research on Cancer WHO classification of tumours of endocrine organs, 4th edition. International Agency for Research on Cancer, Lyon
29.
Zurück zum Zitat Nakamura Y, Larsson C, Julier C, Byström C, Skogseid B, Wells S et al (1989) Localization of the genetic defect in multiple endocrine neoplasia type 1 within a small region of chromosome 11. American Journal of Human Genetics 44:751–755PubMedPubMedCentral Nakamura Y, Larsson C, Julier C, Byström C, Skogseid B, Wells S et al (1989) Localization of the genetic defect in multiple endocrine neoplasia type 1 within a small region of chromosome 11. American Journal of Human Genetics 44:751–755PubMedPubMedCentral
34.
Zurück zum Zitat Szabó J, Heath B, Hill VM, Jackson CE, Zarbo RJ, Mallette LE et al (1995) Hereditary hyperparathyroidism-jaw tumor syndrome: The endocrine tumor gene HRPT2 maps to chromosome 1q21-q31. American Journal of Human Genetics 56:944–950PubMedPubMedCentral Szabó J, Heath B, Hill VM, Jackson CE, Zarbo RJ, Mallette LE et al (1995) Hereditary hyperparathyroidism-jaw tumor syndrome: The endocrine tumor gene HRPT2 maps to chromosome 1q21-q31. American Journal of Human Genetics 56:944–950PubMedPubMedCentral
36.
Zurück zum Zitat Teh BT, Farnebo F, Kristoffersson U, Sundelin B, Cardinal J, Axelson R et al (1996) Autosomal dominant primary hyperparathyroidism and jaw tumor syndrome associated with renal hamartomas and cystic kidney disease: linkage to 1q21-q32 and loss of the wild type allele in renal hamartomas. The Journal of Clinical Endocrinology and Metabolism 81:4204–4211. https://doi.org/10.1210/jcem.81.12.8954016CrossRefPubMed Teh BT, Farnebo F, Kristoffersson U, Sundelin B, Cardinal J, Axelson R et al (1996) Autosomal dominant primary hyperparathyroidism and jaw tumor syndrome associated with renal hamartomas and cystic kidney disease: linkage to 1q21-q32 and loss of the wild type allele in renal hamartomas. The Journal of Clinical Endocrinology and Metabolism 81:4204–4211. https://​doi.​org/​10.​1210/​jcem.​81.​12.​8954016CrossRefPubMed
41.
43.
Zurück zum Zitat Bricaire L, Odou M-F, Cardot-Bauters C, Delemer B, North M-O, Salenave S et al, on behalf of the GTE Group (2013) Frequent large germline HRPT2 deletions in a French National cohort of patients with primary hyperparathyroidism. J Clin Endocrinol Metab 98:E403–408. https://doi.org/10.1210/jc.2012-2789 Bricaire L, Odou M-F, Cardot-Bauters C, Delemer B, North M-O, Salenave S et al, on behalf of the GTE Group (2013) Frequent large germline HRPT2 deletions in a French National cohort of patients with primary hyperparathyroidism. J Clin Endocrinol Metab 98:E403–408. https://​doi.​org/​10.​1210/​jc.​2012-2789
56.
Zurück zum Zitat Uchino S, Noguchi S, Nagatomo M, Sato M, Yamashita H, Yamashita H et al (2000) Absence of somatic RET gene mutation in sporadic parathyroid tumors and hyperplasia secondary to uremia, and absence of somatic Men1 gene mutation in MEN2A-associated hyperplasia. Biomed Pharmacother 54 Suppl 1:100s–103s. https://doi.org/10.1016/s0753-3322(00)80023-2CrossRefPubMed Uchino S, Noguchi S, Nagatomo M, Sato M, Yamashita H, Yamashita H et al (2000) Absence of somatic RET gene mutation in sporadic parathyroid tumors and hyperplasia secondary to uremia, and absence of somatic Men1 gene mutation in MEN2A-associated hyperplasia. Biomed Pharmacother 54 Suppl 1:100s–103s. https://​doi.​org/​10.​1016/​s0753-3322(00)80023-2CrossRefPubMed
78.
Zurück zum Zitat McGregor DH, Lotuaco LG, Rao MS, Chu LL (1978) Functioning oxyphil adenoma of parathyroid gland. An ultrastructural and biochemical study. Am J Pathol 92:691–711PubMedPubMedCentral McGregor DH, Lotuaco LG, Rao MS, Chu LL (1978) Functioning oxyphil adenoma of parathyroid gland. An ultrastructural and biochemical study. Am J Pathol 92:691–711PubMedPubMedCentral
81.
85.
Zurück zum Zitat Hahn MA, Howell VM, Gill AJ, Clarkson A, Weaire-Buchanan G, Robinson BG et al (2010) CDC73/HRPT2 CpG island hypermethylation and mutation of 5’-untranslated sequence are uncommon mechanisms of silencing parafibromin in parathyroid tumors. Endocr Relat Cancer 17:273–282. https://doi.org/10.1677/ERC-09-0291CrossRefPubMed Hahn MA, Howell VM, Gill AJ, Clarkson A, Weaire-Buchanan G, Robinson BG et al (2010) CDC73/HRPT2 CpG island hypermethylation and mutation of 5’-untranslated sequence are uncommon mechanisms of silencing parafibromin in parathyroid tumors. Endocr Relat Cancer 17:273–282. https://​doi.​org/​10.​1677/​ERC-09-0291CrossRefPubMed
109.
Zurück zum Zitat Yu W, McPherson JR, Stevenson M, van Eijk R, Heng HL, Newey P et al (2015) Whole-exome sequencing studies of parathyroid carcinomas reveal novel PRUNE2 mutations, distinctive mutational spectra related to APOBEC-catalyzed DNA mutagenesis and mutational enrichment in kinases associated with cell migration and invasion. J Clin Endocrinol Metab 100:E360-364. https://doi.org/10.1210/jc.2014-3238CrossRefPubMed Yu W, McPherson JR, Stevenson M, van Eijk R, Heng HL, Newey P et al (2015) Whole-exome sequencing studies of parathyroid carcinomas reveal novel PRUNE2 mutations, distinctive mutational spectra related to APOBEC-catalyzed DNA mutagenesis and mutational enrichment in kinases associated with cell migration and invasion. J Clin Endocrinol Metab 100:E360-364. https://​doi.​org/​10.​1210/​jc.​2014-3238CrossRefPubMed
117.
Zurück zum Zitat Juhlin CC, Haglund F, Villablanca A, Forsberg L, Sandelin K, Bränström R et al (2009) Loss of expression for the Wnt pathway components adenomatous polyposis coli and glycogen synthase kinase 3-beta in parathyroid carcinomas. Int J Oncol 34:481–492PubMed Juhlin CC, Haglund F, Villablanca A, Forsberg L, Sandelin K, Bränström R et al (2009) Loss of expression for the Wnt pathway components adenomatous polyposis coli and glycogen synthase kinase 3-beta in parathyroid carcinomas. Int J Oncol 34:481–492PubMed
124.
Zurück zum Zitat Lubensky IA, Debelenko LV, Zhuang Z, Emmert-Buck MR, Dong Q, Chandrasekharappa S et al (1996) Allelic deletions on chromosome 11q13 in multiple tumors from individual MEN1 patients. Cancer Research 56:5272–5278PubMed Lubensky IA, Debelenko LV, Zhuang Z, Emmert-Buck MR, Dong Q, Chandrasekharappa S et al (1996) Allelic deletions on chromosome 11q13 in multiple tumors from individual MEN1 patients. Cancer Research 56:5272–5278PubMed
133.
134.
138.
Zurück zum Zitat Vargas MP, Vargas HI, Kleiner DE, Merino MJ (1997) The role of prognostic markers (MiB-1, RB, and bcl-2) in the diagnosis of parathyroid tumors. Mod Pathol 10:12–17PubMed Vargas MP, Vargas HI, Kleiner DE, Merino MJ (1997) The role of prognostic markers (MiB-1, RB, and bcl-2) in the diagnosis of parathyroid tumors. Mod Pathol 10:12–17PubMed
141.
Zurück zum Zitat Vasef MA, Brynes RK, Sturm M, Bromley C, Robinson RA (1999) Expression of cyclin D1 in parathyroid carcinomas, adenomas, and hyperplasias: a paraffin immunohistochemical study. Mod Pathol 12:412–416PubMed Vasef MA, Brynes RK, Sturm M, Bromley C, Robinson RA (1999) Expression of cyclin D1 in parathyroid carcinomas, adenomas, and hyperplasias: a paraffin immunohistochemical study. Mod Pathol 12:412–416PubMed
152.
Metadaten
Titel
Genomics and Epigenomics in Parathyroid Neoplasia: from Bench to Surgical Pathology Practice
verfasst von
C. Christofer Juhlin
Lori A. Erickson
Publikationsdatum
02.12.2020
Verlag
Springer US
Erschienen in
Endocrine Pathology / Ausgabe 1/2021
Print ISSN: 1046-3976
Elektronische ISSN: 1559-0097
DOI
https://doi.org/10.1007/s12022-020-09656-9

Weitere Artikel der Ausgabe 1/2021

Endocrine Pathology 1/2021 Zur Ausgabe

Neu im Fachgebiet Pathologie