Abstract
The classification of human cancers represents one of the cornerstones of modern pathology. Over the last century, surgical pathologists established the current taxonomy of neoplasia using traditional histopathological parameters, which include tumor architecture, cytological features and cellular proliferation. This morphological classification is efficient and robust with high reproducibility and has served patients and health care providers well. The most recent decade has witnessed an explosion of genome-wide molecular genetic and epigenetic data for most cancers, including tumors of endocrine organs. The availability of this expansive multi-dimensional genomic data, collectively termed the cancer genome, has catalyzed a re-examination of the classification of endocrine tumors. Here, recent cancer genome studies of various endocrine tumors, including those of the thyroid, pituitary and adrenal glands, pancreas, small bowel, lung and skin, are presented with special emphasis on how genomic insights are impacting endocrine tumor classification.
Introduction
Human cancers are genomic diseases characterized by germline and somatic genetic defects that drive and define the neoplastic phenotype (Stratton et al. 2009), which includes the ability of transformed cells to invade adjacent normal tissues and metastasize to distant sites (Hanahan & Weinberg 2011). Identifying and understanding the cellular and molecular consequences of these mutations has been one of the areas of intense focus in cancer research for the last several decades. These efforts were greatly accelerated by sequencing the human genome (Lander et al. 2001, Venter et al. 2001). Subsequently, during the most recent decade, the genomes of the most common types of human cancer have been elucidated and characterized, largely due to remarkable advances in next-generation sequencing technologies (Metzker 2010, Goodwin et al. 2016, Levy & Myers 2016). Coordinated cancer genome discovery and characterization efforts by multi-disciplinary networks of investigators, such as The Cancer Genome Atlas (TCGA; https://cancergenome.nih.gov) (McCain 2006, Cancer Genome Atlas Research 2013, Wang et al. 2016) and the International Cancer Genome Consortium (ICGC; https://icgc.org) (Zhang et al. 2011, Jennings & Hudson 2013), as well as numerous institutional studies, have systematically addressed most common cancers, as well as some rare types. Collectively, these integrated, multi-dimensional genomic studies have permitted a re-evaluation of human cancer at the most fundamental level, i.e. pathologic tumor classification, which has been traditionally based on histopathology. In many tumor types, these studies have confirmed existing tumor taxonomies, which in fact are the expected results since the morphology of a given tumor reflects its cumulative genetic and epigenetic changes. However, in most tumors, analysis of genomic data has revealed previously unrecognized molecular heterogeneity within pathologic entities and catalyzed significant refinements of tumor classification schemes. Together with the identification of novel genetic alterations, these results have profound implications for the treatment of cancer patients.
Tumors of endocrine organs are similarly genomic diseases. Compared to more common cancers, endocrine tumors sometimes do not attract equal attention and resources largely due to their relative rare nature. Fortunately for these patients, many endocrine cancer types have been investigated as part this genomic revolution. Here, the primary genomic studies of endocrine tumors will be reviewed, with special emphasis on those studies with significant taxonomic implications.
Power of integrated genomic analysis
Initially, the majority of genome-wide genetic and epigenetic studies interrogated a single molecular component of the cancer genome, such as the transcriptome, methylome or compendium of copy number alterations. While these single-platform studies have successfully advanced our knowledge of many cancer types, the availability of multi-platform molecular data on the same tumor cohort permits a higher level of analytical integration, which yields greater understanding of tumorigenesis and enhanced hypothesis generation. For example, the TCGA thyroid cancer study (Cancer Genome Atlas Research Network 2014) confirmed increased expression of an oncogenic microRNA (mir-21) in tumors with BRAF V600E mutation and aggressive histologic features. However, by incorporating DNA methylation data, which revealed altered methylation of the mir-21 promoter, a hypothesis was generated in which epigenetic regulation of microRNAs plays a role in the development of aggressive forms of BRAF V600E-mutated thyroid cancer. Another such example from the TCGA adrenal cortical carcinoma project (Zheng et al. 2016) combined telomere length data with genotype, gene expression and copy number data to extract critical insights into the role of telomere maintenance and whole-genome doubling in these cancers. Such sophisticated bioinformatic analyses are not possible when only single-platform molecular data are available. As such, the true power of the TCGA, ICGA and similar integrated genomic characterization studies lies in their ability to leverage multi-dimensional genomic data to derive novel insights into the molecular underpinnings and classification of cancer.
Thyroid cancer
Follicular cell-derived carcinomas
Tumors of the thyroid gland derived from follicular cells are broadly divided into differentiated and undifferentiated subtypes. Differentiated thyroid cancer includes papillary thyroid carcinoma (PTC) and its many histological variants, follicular thyroid carcinoma (FTC) and Hurthle cell (oncocytic) carcinoma. The remaining tumors are classified as either poorly differentiated thyroid carcinoma (PDCA) or anaplastic thyroid carcinoma (ATC) depending on their histology and degree of thyroid-related gene expression. By definition, ATC has essentially lost all of its follicular cell differentiation; hence, its alternative designation of undifferentiated thyroid carcinoma. Despite the prognostic significance of the differentiated vs undifferentiated taxonomy, it is now fully recognized that such a coarse classification scheme is inadequate to capture the full clinicopathologic and genetic diversity seen across thyroid cancers.
PTC represents the most common follicular cell thyroid cancer (Kitahara & Sosa 2016). As such, it was selected for study by TCGA for its project on thyroid cancer. The study cohort included 496 primary tumors analyzed using their standardized molecular platforms that included whole exome DNA sequencing, RNA sequencing, microRNA sequencing, copy number analysis, DNA methylation profiling and proteomic profiling (Cancer Genome Atlas Research Network 2014, Giordano 2014). For the sake of simplicity, PTCs were histologically classified into one of three main subtypes: classical type, tall cell variant and follicular variant.
Despite the fact that PTC is a relatively indolent cancer with an excellent overall prognosis and survival, many insights were nonetheless derived from its genomic characterization (Fig. 1). PTC was observed to have an overall low tumor mutational burden (Table 1) and a stable genome with few copy number alterations. Prevalent somatic alterations included BRAF V600E, RET and other tyrosine kinase gene fusions and RAS family point mutations. These mutations were almost entirely mutually exclusive, which strongly suggests that possessing more one of these driver mutations does not provide a biological advantage. The mutual exclusive nature of BRAF V600E and RAS mutations permitted the development of a BRAF V600E-RAS score, termed BRS, which was derived from distinct gene expression profiles of a selected 71-gene set. The BRS was used as a framework to visualize the entirety of the genomic data and interrogate the biology of the non-BRAF V600E and non-RAS somatic mutations (Fig. 2). Using this approach, PTCs were divided into two broad groups, termed BRAF V600E-like and RAS-like. This overarching result illustrates the striking biological differences between BRAF V600E-like and RAS-like PTCs. This result correlated to a high degree with tumor morphology, in which BRAF V600E-like PTCS are pathologically characterized by the presence of true papillae (classical type PTC) or tall cell features (tall cell variant PTC) and, conversely, RAS-like PTCs are distinguished by a pure follicular morphology without papillae or tall cell features (follicular variant PTC). Beyond the BRAF V600E-like and RAS-like distinction, it became possible to further divide the BRAF V600E-like group into two groups defined by BRAF V600E and RET plus other gene fusions. While these observations match well with numerous existing studies (Zhu et al. 2003, Adeniran et al. 2006, Brzezianska et al. 2007, Koperek et al. 2012), the size of the TCGA cohort and the strength and scope of the resulting data were enough to catalyze a deeper recognition and understanding of the different histologic and molecular subtypes of PTC. These insights have broad implications for pathology practice, clinical trial design and routine clinical care (Fagin & Wells 2016).
Tumor mutational burden across the spectrum of endocrine cancers.
Endocrine tumor type | Tumor mutational burden, mean | Tumor mutational burden, range | References |
---|---|---|---|
Mean, non-silent mutations per MB | Range, non-silent mutations per MB | ||
Papillary thyroid carcinoma | 0.41 | 0.00–2.158 | Cancer Genome Atlas Research Network (2014) |
Poorly differentiated thyroid carcinoma | >PTC | Unknown | Landa et al. (2016) |
Anaplastic thyroid carcinoma | >>>PTC | Unknown | Landa et al. (2016) |
Pituitary adenoma | 0.6 | 0.14–4.42 | Bi et al. (2017b) |
Pancreatic NETs | 0.82 | 0.04–4.56 | Scarpa et al. (2017) |
Small intestinal NETs | 0.77 | 0.13–2.51 | Francis (2013) |
Adrenal cortical carcinoma | 0.9 | 0.2–14.0 | Zheng et al. (2016) |
Pheochromocytoma/paraganglioma | Low | Unknown | Fishbein et al. (2017) |
Small cell lung carcinoma | 8.62 | Unknown | George et al. (2015) |
Merkel cell carcinoma, MCPyV negative | 10.09 | Unknown | Harms et al. (2015) |
Merkel cell carcinoma, MCPyV positive | 0.4 | Unknown | Harms et al. (2015) |
The overarching conclusion from the TCGA study raises the fundamental question of whether BRAF V600E-like and RAS-like PTCs belong together as a single diagnostic entity. One alternative classification would be to reclassify RAS-like PTCs as FTCs because of their shared follicular growth pattern and predominance of RAS mutations (Esapa et al. 1999, Gupta et al. 2013, Medici et al. 2015). The significance of such a classification revision would impact many aspects of thyroid cancer care, including risk assessment and use of radioiodine (Asa et al. 2015).
FTCs have also been genomically investigated, but to a lesser degree given their rare incidence compared to PTC. A recent study of follicular adenomas and FTCs revealed similar tumor mutational burdens and evolutionary ages, suggesting a close pathogenetic relationship (Jung et al. 2016). This result is generally consistent with another recent study of minimally invasive, angioinvasive and widely invasive FTCs that demonstrated similar genomic features, including tumor mutational burden. Despite this similarity, mutational burden was an independent predictor of structural recurrence and mortality (Nicolson et al. 2018).
The newly described follicular cell entity termed noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP) defines a pure follicular neoplasm that is encapsulated or circumscribed without capsular or lymphovascular invasion and displays the nuclear features of papillary carcinoma (Nikiforov et al. 2016). NIFPTs, when properly diagnosed, have an exceptionally low risk of structural recurrence and accordingly were proposed as a new diagnostic entity to remove ‘carcinoma’ from the diagnostic nomenclature, thereby addressing in part the issue of thyroid cancer overdiagnosis (Brito & Hay 2017). NIFTPs are predominantly characterized by RAS mutations and therefore are considered to be RAS-like tumors (Paulson et al. 2017, Zhao et al. 2017, Kim et al. 2018b ). Some preliminary gene expression data suggest that NIFTPs can exist in two forms, one similar to follicular adenomas and another similar to invasive follicular variants of PTC.
Recent genomic studies of PDCA have replicated the BRAF V600E-like and RAS-like distinction observed in PTC, in addition to demonstrating an increased mutational burden (Table 1) (Landa et al. 2016). Regarding this distinction, a strong correlation with the type of PDCA was evident. PDCAs have been previously classified into two distinct histologic types. The Turin-type of PDCA is defined by growth pattern (e.g. insular) and mitotic rate (Volante et al. 2007, 2016). Conversely, the Memorial Sloan Kettering (MSK)-type of PDCA is defined on the basis of necrosis and mitotic rate, without an insular growth pattern (Hiltzik et al. 2006). In the Landa et al. study, RAS-like tumors were predominantly Turin-type (insular) PDCAs, whereas BRAF V600E-like tumors were enriched for MSK-type (non-insular) PDCAs. Thus, the available genomic data support two distinct types of PDCA with strong histological association and distinct patterns of metastasis in which the RAS-like tumors often present with distant metastatic disease, and BRAF V600E-like tumors with regional lymph nodes.
The Landa et al. study also analyzed a cohort of ATC. Collectively, ATCs displayed a higher overall mutational burden (Table 1) compared to PTC and PDCA, with significantly higher rates of TERT promoter mutations. Regarding the BRAF V600E-like and RAS-like distinction observed in PTC and PDCA, all of the ATCs had a BRAF V600E-like phenotype irrespective of their genotype. This result may reflect a combination of a greatly increased tumor mutational burden and the presence of intratumoral inflammatory cells (i.e. macrophages) that together effectively eclipse the BRS gene expression signature. The collective insights derived from these studies are summarized in Fig. 3 (Giordano 2018).
Medullary carcinoma
The genetic landscape of medullary thyroid carcinoma (MTC) is distinct from follicular cell derived thyroid cancers (Ciampi et al. 2017). MTC is divided into heritable (30%) and sporadic (70%) forms and is characterized by germline and somatic point mutations of the RET proto-oncogene, respectively (Accardo et al. 2017). RET-negative MTCs contain RAS mutations (Ciampi et al. 2013) and rare RET fusions (Grubbs et al. 2015). Rare ALK fusions have also been identified by genomic studies (Ji et al. 2015). Recent immunohistochemical work suggests that loss of RB is associated with decreased survival (Valenciaga et al. 2017). To date, an integrated, multi-dimensional genomic characterization of MTC that sheds light on tumor classification has not been reported.
Pituitary adenoma and carcinoma
Genomic evaluation of pituitary adenomas, the most common CNS neoplasm (Theodros et al. 2015), is being deployed to refine their histological and molecular classification (Bi et al. 2018). Sporadic pituitary adenomas, similar to other low-grade endocrine tumors, are characterized by an overall low somatic tumor mutational burden (Table 1). Recurrently mutated genes in pituitary adenomas are enriched in a small number of pathways: cell cycle (PIK3CA, NOTCH1/2, and GLI1/2/3), chromatic modification and transcriptional regulation (ARID1A/B, ASXL1, BRD4 and CREBBP) and DNA damage response (PRKDC, BRCA1/2, ATM and FANCA) (Bi et al. 2017a ). Mutational patterns differ according to pituitary adenoma functional subtype, with mutations of the deubiquitinase USP8 found in up to two-thirds of corticotroph adenomas (Ma et al. 2015, Reincke et al. 2015). Somatotroph adenomas contain frequent GNAS mutations (Ronchi et al. 2016, Bi et al. 2017a ) and GPR101 E308D is found in a small number of acromegaly patients (Trivellin et al. 2014). MEN1 mutations are found in both familiar and sporadic cases (Uraki et al. 2017). Null cell adenomas have few recurrent point mutations, suggesting a role for alterative tumorigenic mechanisms such as copy number changes, gene fusions and/or epigenetic alterations. Indeed, chromosomal copy number changes have been identified and divide pituitary adenomas into two classes: a quiet group with few changes and a disrupted group with extensive arm-level genomic alterations (Bi et al. 2017a ,b ). Disrupted adenomas were associated with functional status, although there was no association between copy number status and atypical histology, Ki-67 proliferation index and recurrence (Bi et al. 2017a ). Further clinical significance of this chromosomal classification awaits validation with larger cohorts.
Epigenetic changes are frequent in pituitary adenomas and exhibit subtype specificity (Farrell 2014). Epigenetic regulation may play a role in tumor cell invasion (Gu et al. 2016), although there is a lack of agreement between studies. A recent DNA methylation profiling study of null cell adenomas confirmed dysregulation of key pathways, although differences between invasive and noninvasive tumors were minimal (Kober et al. 2018).
Exploration of the cancer genome of pituitary carcinoma has been limited by the extremely rare nature of this neoplasm (Yang et al. 2016). Pituitary carcinomas have been reported in patients with Lynch syndrome (Bengtsson et al. 2017) and SDHB mutations (Tufton et al. 2017), but an integrated genomic analysis of these tumors has not been reported.
Adrenal tumors
Adrenal cortical tumors
Adrenal cortical tumors are pathologically classified as adrenal cortical adenomas when they lack malignant potential, whereas those tumors that possess malignant potential are diagnosed as adrenal cortical carcinoma (ACC). The pathological assessment of malignant potential is straightforward for the majority of tumors using established clinical and histopathological parameters that are often combined into one of several diagnostic systems (e.g. Weiss system (Weiss 1984)). Occasional tumors defy classification and can be diagnosed as adrenal cortical tumors of uncertain malignant potential.
Genomic information has greatly expanded the understanding of adrenal cortical tumors, which has led to increased personalized care of patients with ACC (Assie et al. 2014a ). Moreover, molecular data derived from genomic studies generally correlate with histological parameters, which has resulted in advances in routine pathological assessment of these tumors.
In the course of developing the above-mentioned diagnostic systems, it was recognized that tumoral mitotic rate could be the basis for a proliferation-based prognostic grading scheme, classifying ACCs as either low or high grade (Weiss et al. 1989). This simple two-class scheme was confirmed by studies of the cancer transcriptome using independent patient cohorts (de Reynies et al. 2009, Giordano et al. 2009) and has been largely accepted into routine pathology practice (Giordano 2011). Along similar proliferation-related lines, the Ki-67 proliferation index assessed by routine immunohistochemical staining has also been shown to have major prognostic significance (Morimoto et al. 2008, Beuschlein et al. 2015) and has also been recommended for routine practice via the most recent College of American Pathologists’ protocol for examination of adrenal cortical tumors.
An integrated genomic characterization of 45 ACCs by the European Network for the Study of Adrenal Tumors (ENSAT) (Assie et al. 2014b ) identified novel genetic alterations (e.g. ZNRF3, DAXX and TERT) and provided strong support for a two-group classification with different patient outcomes. The poor outcome group, termed C1A, had a higher mutation burden and increased DNA methylation changes. In contrast, the good outcome group, termed C1B, had fewer mutations and was characterized by deregulation of microRNA clusters. Beyond the two-class system, the data suggested that methylation status might further divide the C1A group into different prognostic subgroups.
TCGA performed a similar integrated, multi-platform genomic analysis using a near-global cohort of 91 ACCs (Zheng et al. 2016), which confirmed many of the prior findings from the ENSAT study. This study identified a relatively higher tumor mutational burden (Table 1) and further extended the cohort of ACC driver mutations to include PRKARI1, RPL22, TERF2, CCNE1 and NF1 and illustrated the dominant role that copy number alterations, including whole-genome doubling, play in ACC pathogenesis and progression. Regarding ACC classification, three distinct outcome groups were identified using a cluster of cluster (COC) analysis of the complete set of genomic data (Fig. 4), which could be nearly replicated by a 68-CpG probe methylation signature. This COC solution also provided a context for the increased understanding of the different types of ACC. COC1 tumors largely corresponded to C1B tumors in the ENSAT study and had distinct genomic (e.g. transcriptional) profiles with a lower tumor mutational burden. COC2 and COC3 tumors corresponded to C1A tumors in the ENSAT study and differed in transcriptional, methylation and copy number profiles. The TCGA study effectively divided the C1A group in the ENSAT study into two distinct prognostic subgroups. These collective findings are illustrated in Fig. 5.
Translation of this three-group ACC classification via an assay that can be performed with routinely available pathology materials, such as formalin-fixed paraffin-embedded tissue, is currently underway by several groups. The successful implementation of a routine clinical assay will represent a meaningful advance for the classification of ACCs by providing a more refined assessment of tumor biology and prognosis, with the potential to impact patient care.
Pheochromocytoma and paraganglioma
Tumors of the adrenal medulla and extra-adrenal paraganglia are highly related tumors with similar histological and genetic features. Pheochromocytoma is defined by the World Health Organization (WHO) as ‘a tumor of chromaffin cells that arises in the adrenal medulla’, whereas extra-adrenal paragangliomas are ‘tumors originating from neural crest-derived paraganglion cells situated in the region of the autonomic nervous system ganglia and accompanying nerves’ (Lloyd et al. 2017).
Pheochromocytomas and paragangliomas (PPGL) have a very high degree of heritability (up to 40% of patients) and are part of many hereditary syndromes (Dahia 2014, Favier et al. 2015). Innumerable studies have identified the germline and somatic mutations that drive tumorigenesis (Turchini et al. 2018). However, the TCGA genomic characterization of PPGL extended the knowledge base of these tumors in several aspects, including the identification of novel mutations such as recurrent MAML3 gene fusions and CSDE1 frameshift and splice-site mutations (Fishbein et al. 2017). These mutations display a high degree of mutual exclusivity (Fig. 6), as observed in other endocrine tumors with relatively low mutational burdens, demonstrating a lack of clonal advantage of having more than one such mutation. Relevant to tumor classification, this study expanded the molecular taxonomy of PPGL by identifying a Wnt signaling group of tumors in addition to confirming the previously described molecular classes termed pseudohypoxia (Jochmanova et al. 2015) and kinase signaling (Bjorklund et al. 2016). This three-group classification has significant implications for many aspects of PPGL diagnosis, risk assessment and treatment, as summarized in Fig. 7.
Neuroblastoma
Neuroblastoma belongs to a group of neuroblastic tumors that arise from the sympathoadrenal lineage of the neural crest. These tumors represent a wide range of tumor biology that spans from benign (ganglioneuroma) to malignant tumors (ganglioneuroblastoma and neuroblastoma). Neuroblastoma is characterized by variable clinical behavior and response to therapy. Decades ago, it was discovered that amplification of the MYCN oncogene identified a minority of neuroblastomas with high-risk disease (Schwab et al. 1984), and MYCN amplification status remains a clinically relevant biomarker (Westermark et al. 2011). Beyond MYCN alterations, other somatic mutations are relatively rare (Molenaar et al. 2012, Pugh et al. 2013) and include ALK amplification and point mutations of ALK, ATRX, PTPN11, TIAM1 and ARIA1A/B (Sausen et al. 2013, Bresler et al. 2014).
Genome-wide evaluation of chromosomal copy number alterations confirmed and extended the MYCN-based molecular classification (Janoueix-Lerosey et al. 2009). Similar to ACCs, tumors characterized with arm-level chromosomal changes were associated with a favorable outcome compared to high-risk tumors that possessed more segmental alterations, with an observed correlation between outcome and the number of segmental alterations (Schleiermacher et al. 2010).
Transcriptome studies of neuroblastoma have identified several prognostic gene expression signatures that are independent of established prognostic factors (Fardin et al. 2010, De Preter et al. 2011, Asgharzadeh et al. 2012, Applebaum et al. 2016), and some profiles using routinely available tissue sample are moving toward clinical implementation (Stricker et al. 2014). Collectively, these studies and others highlight some of the translational advances that are leading to improved molecular risk stratification and potential novel therapeutic approaches (Bosse & Maris 2016).
Pancreatic neuroendocrine tumors
The genomic landscape of pancreatic neuroendocrine (NE) neoplasms (PanNENs) has been significantly elucidated by recent studies (Jiao et al. 2011, Scarpa et al. 2017, Wong et al. 2017, Mafficini & Scarpa 2018). Genetically, PanNENs are markedly distinct from more common primary pancreatic cancers, such as adenocarcinoma (Jiao et al. 2011, Cancer Genome Atlas Research 2017).
PanNENs are broadly divided into well- and poorly differentiated groups, termed PanNETs and PanNECs, respectively. The WHO further classifies them into three grades (G1, G2 and G3). PanNETs are graded into G1, G2 or G3 groups based on proliferation, whereas PanNECs are uniformly graded as G3 tumors (Lloyd et al. 2017). PanNETs and PanNECs have significantly different genetic profiles, consistent with their distinct clinical behavior. PanNETs possess an indolent growth and have a relatively low tumor mutational burden (Table 1) (Scarpa et al. 2017). Beyond some of the initial genes to be identified in PanNETs, such as MEN1 (Gortz et al. 1999), genomic studies have expanded the universe of somatic driver genes to include DAXX, ATRX, TSC1/2, PTEN, NF1, DEPDC5 and EWSR1 gene fusions (Jiao et al. 2011, Scarpa et al. 2017). Classification of PanNETs based on mutational profiles has identified four core pathway-related groups: DNA damage repair, chromatin remodeling, mTOR signaling and altered telomere maintenance. Regarding prognostic grading, some of these PanNETs classes have been associated with distinct clinical behavior. Overall, PanNETs with MEN1 and DAXX/ATRX mutations have a better prognosis (Jiao et al. 2011), although intermediate-grade (G2) PanNETs with DAXX/ATRX mutations have a poor prognosis within this grade group (Scarpa et al. 2017). Tumors without altered telomere length were associated with a better outcome (Scarpa et al. 2017). While these associations need further validation with larger cohorts, the collective data suggest that mutational status possesses significant prognostic information for PanNETs.
In addition to the pathway-based classification of PanNETs, it was possible to identify five types of tumors based on mutational signatures (Scarpa et al. 2017), including a novel signature termed MUTYH inactivation because of biallelic inactivation of MUTYH (germline-inactivating mutations combined with simultaneous deletion of the other allele). Other observed signatures included BRCA, APOBEC and AGE. It is clear from the collective genomic data that PanNETs, as a group, contain significantly more molecular heterogeneity than predicted based on histology alone.
PanNECs have distinct genetic profiles from PanNETs, with frequent mutations in TP53, RB1 and CDKN2A and DAXX/ATRX mutations are notably absent in the high-grade carcinomas regardless of their small-cell or large cell histology (Yachida et al. 2012). These genetic differences support the classification of PanNECs as high-grade (G3) neoplasms, defined on the basis of proliferation activity (>20 mitoses per 10 hpfs or a Ki-67 proliferation index of >20%) in the WHO classification of PanNENs (Lloyd et al. 2017).
Small intestinal neuroendocrine tumors
Neuroendocrine tumors of the small intestine (SINETs) represent the most common small intestinal cancer and are increasing in incidence (Yao et al. 2008). Similar to other low-grade endocrine tumors, genomic studies have revealed a relatively low mutational burden (Table 1) (Banck et al. 2013, Francis et al. 2013). Genomic studies have identified few recurrent alterations, including loss of heterozygosity at chromosome 18 (Cunningham et al. 2011), arm-level chromosomal gains (Kulke et al. 2008) and mutations of CDKN1B (Francis et al. 2013). An integrated genomic study of SINETs revealed significant molecular diversity within SINETs and divided tumors into three groups based on CDKN1B mutational status, epigenetic profiles and copy number profiles (Karpathakis et al. 2016). Importantly, these molecular groups were associated with distinct survival. This provocative study awaits validation and translation to routine practice.
Lung neuroendocrine tumors
Neuroendocrine tumors of the lung represent a biologically diverse set of related tumors that are distinct from non-small-cell lung carcinomas, such as adenocarcinoma and squamous cell carcinoma. Neuroendocrine tumors are classified into three main groups based on histological features. Low-grade neuroendocrine tumors, termed typical carcinoids (TCs) and intermediate-grade neuroendocrine tumors, termed atypical carcinoids (ACs) are histologically and molecularly distinct from high-grade neuroendocrine tumors, which are divided into small-cell (SCLC) and large-cell (LCLC) types. The collective molecular data support this existing histological classification, with carcinoids and carcinomas displaying distinct mutational burdens and profiles (Swarts et al. 2012, Simbolo et al. 2017). For example, somatic TP53 mutations and loss of 3p are common in NE carcinomas, yet rare in carcinoids (Rossi et al. 2018). These molecular differences are directly related to differences in tobacco exposure, which is known to cause TP53 and other mutations (Gibbons et al. 2014). In contrast, MEN1 mutations are nearly exclusively present in carcinoids (Simbolo et al. 2017).
A genomic study of a large cohort of small-cell carcinomas revealed a high mutational burden with bi-allelic inactivation of TP53 and RB in nearly all cases, suggesting that loss of these tumor suppressors is an obligatory event in this tumor (George et al. 2015). In addition, kinase mutations and NOTCH family mutations were frequent. Unsupervised hierarchical clustering of transcriptome data identified two classes with distinct gene expression profiles of various NE markers, yet similar mutational profiles. These classes reflected differing levels of NOTCH pathway activation and may form the basis for a future molecular classification of SCLC. This comprehensive study, supported by mouse studies, provides evidence that NOTCH functions as a tumor suppressor and master regulator of NE differentiation in SCLC.
Skin neuroendocrine tumors
Primary cutaneous neuroendocrine carcinoma, termed Merkel cell carcinoma (MCC), is a rare, clinically aggressive neoplasm with a high mortality rate (Llombart et al. 2017). The incidence of MCC has increased dramatically in recent decades, especially in elderly patients with significant sun exposure. Histologically, it shares features with small-cell carcinoma of the lung, although some MCC contain a mixed non-neuroendocrine component such as squamous cell carcinoma. Molecularly, MCC can be divided into two etiologically distinct groups. One group is characterized by viral integration of Merkel cell polyomavirus that contributes to MCC tumorigenesis by suppressing RB via inhibition by large T antigen (Feng et al. 2008). In contrast, the other group lacks Merkel cell polyomavirus and is characterized by UV light-mediated mutations, such as frequent TP53, RB1, and NOTCH mutations. These two MCC classes are biologically and clinically distinct, with differing tumor mutational burdens (Table 1) and landscapes (Harms et al. 2015, Goh et al. 2016), copy number profiles (Paulson et al. 2009), and outcomes, with viral cases carrying a better prognosis. The collective evidence suggests that these two MCC groups arise from different cells, with viral-negative and viral-positive tumors arising from epidermal keratinocytes and dermal fibroblasts (Harms et al. 2015), respectively, although this awaits validation.
Significance of mutational burden
From the collective available information on tumor mutational burden across endocrine tumors (Table 1), a clear relationship between tumor mutational burden and tumor behavior emerges. While this relationship is generally found across all cancers, and tumor mutational burden is increasingly recognized as an important genomic metric (Chalmers et al. 2017) that can predict response to immunotherapy (Goodman et al. 2017, Yarchoan et al. 2017, Steuer & Ramalingam 2018), it may be especially relevant and revealing in endocrine neoplasia (Hellmann et al. 2018, Kim et al. 2018a ). Many genomic assays have incorporated measurements of tumor mutational burden, and it will be interesting to see what role it might play in endocrine tumor evaluation and patient care in the coming years.
Therapeutic implications of genomic characterization of endocrine tumors
Advances in genomic characterization of human cancers have identified 12 core signaling pathways that serve to regulate essential cellular functions, including cell fate and survival and maintenance of the genome (Vogelstein et al. 2013). The classification of human cancer has the potential to evolve toward a pathway-based taxonomy, thereby supplementing the existing organ-based approach (Sanchez-Vega et al. 2018). This transformation has already begun with the approval of immunotherapies (i.e. pembrolizumab) based on genetic alterations (e.g. mismatch repair defects) independent of tumor origin or histology (Le et al. 2015). This pathway-based approach is being applied to endocrine tumors (e.g. ACC) and will likely accelerate in the coming years, although the existing cell-of-origin taxonomy retained its biological significance in a TCGA pan-cancer analysis of 33 cancer types (Hoadley et al. 2018). This suggests that cell-of-origin context will be a significant factor in therapeutic response to targeted therapy, independent of genotype, signaling pathway activation and other genomic features.
Conclusion
Coordinated genomic discovery programs like TCGA and ICGC are characterizing the cancer genome for many cancer types, including endocrine tumors. The last decade has seen significant accomplishments that can be broadly divided into improved understanding of the molecular basis of cancer, revised tumor classification schemes through discovery of novel molecular subtypes, and identification of genomic attributes of tumors that can serve as therapeutic targets and predictors of response. The availability of robust molecular classification schemes for endocrine cancers is leading to advances in the diagnosis, prognostication and treatment of these patients.
Declaration of interest
The author declares that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.
Funding
This work did not receive any specific grant from any funding agency in the public, commercial, or not-for-profit sector.
Acknowledgements
The author would like to thank my colleagues in the Department of Pathology for their support, members of my laboratory for their hard work, and the guest editors of this section for the opportunity to contribute to this special issue.
References
Accardo G, Conzo G, Esposito D, Gambardella C, Mazzella M, Castaldo F, Di Donna C, Polistena A, Avenia N, Colantuoni V, et al. 2017 Genetics of medullary thyroid cancer: an overview. International Journal of Surgery 41 (Supplement 1) S2–S6. (https://doi.org/10.1016/j.ijsu.2017.02.064)
Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, Biddinger PW & Nikiforov YE 2006 Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. American Journal of Surgical Pathology 30 216–222. (https://doi.org/10.1097/01.pas.0000176432.73455.1b)
Applebaum MA, Jha AR, Kao C, Hernandez KM, DeWane G, Salwen HR, Chlenski A, Dobratic M, Mariani CJ, Godley LA, et al. 2016 Integrative genomics reveals hypoxia inducible genes that are associated with a poor prognosis in neuroblastoma patients. Oncotarget 7 76816–76826. (https://doi.org/10.18632/oncotarget.12713)
Asa SL, Giordano TJ & LiVolsi VA 2015 Implications of the TCGA genomic characterization of papillary thyroid carcinoma for thyroid pathology: does follicular variant papillary thyroid carcinoma exist? Thyroid 25 1–2. (https://doi.org/10.1089/thy.2014.0540)
Asgharzadeh S, Salo JA, Ji L, Oberthuer A, Fischer M, Berthold F, Hadjidaniel M, Liu CW, Metelitsa LS, Pique-Regi R, et al. 2012 Clinical significance of tumor-associated inflammatory cells in metastatic neuroblastoma. Journal of Clinical Oncology 30 3525–3532. (https://doi.org/10.1200/JCO.2011.40.9169)
Assie G, Jouinot A & Bertherat J 2014a The ‘omics’ of adrenocortical tumours for personalized medicine. Nature Reviews Endocrinology 10 215–228. (https://doi.org/10.1038/nrendo.2013.272)
Assie G, Letouze E, Fassnacht M, Jouinot A, Luscap W, Barreau O, Omeiri H, Rodriguez S, Perlemoine K, Rene-Corail F, et al. 2014b Integrated genomic characterization of adrenocortical carcinoma. Nature Genetics 46 607–612. (https://doi.org/10.1038/ng.2953)
Banck MS, Kanwar R, Kulkarni AA, Boora GK, Metge F, Kipp BR, Zhang L, Thorland EC, Minn KT, Tentu R, et al. 2013 The genomic landscape of small intestine neuroendocrine tumors. Journal of Clinical Investigation 123 2502–2508. (https://doi.org/10.1172/JCI67963)
Bengtsson D, Joost P, Aravidis C, Askmalm Stenmark M, Backman AS, Melin B, von Salome J, Zagoras T, Gebre-Medhin S & Burman P 2017 Corticotroph pituitary carcinoma in a patient with lynch syndrome (LS) and pituitary tumors in a nationwide LS cohort. Journal of Clinical Endocrinology and Metabolism 102 3928–3932. (https://doi.org/10.1210/jc.2017-01401)
Beuschlein F, Weigel J, Saeger W, Kroiss M, Wild V, Daffara F, Libe R, Ardito A, Al Ghuzlan A, Quinkler M, et al. 2015 Major prognostic role of Ki67 in localized adrenocortical carcinoma after complete resection. Journal of Clinical Endocrinology and Metabolism 100 841–849. (https://doi.org/10.1210/jc.2014-3182)
Bi WL, Greenwald NF, Ramkissoon SH, Abedalthagafi M, Coy SM, Ligon KL, Mei Y, MacConaill L, Ducar M, Min L, et al. 2017a Clinical identification of oncogenic drivers and copy-number alterations in pituitary tumors. Endocrinology 158 2284–2291. (https://doi.org/10.1210/en.2016-1967)
Bi WL, Horowitz P, Greenwald NF, Abedalthagafi M, Agarwalla PK, Gibson WJ, Mei Y, Schumacher SE, Ben-David U, Chevalier A, et al. 2017b Landscape of genomic alterations in pituitary adenomas. Clinical Cancer Research 23 1841–1851. (https://doi.org/10.1158/1078-0432.CCR-16-0790)
Bi WL, Larsen AG & Dunn IF 2018 Genomic alterations in sporadic pituitary tumors. Current Neurology and Neuroscience Reports 18 4. (https://doi.org/10.1007/s11910-018-0811-0)
Bjorklund P, Pacak K & Crona J 2016 Precision medicine in pheochromocytoma and paraganglioma: current and future concepts. Journal of Internal Medicine 280 559–573. (https://doi.org/10.1111/joim.12507)
Bosse KR & Maris JM 2016 Advances in the translational genomics of neuroblastoma: from improving risk stratification and revealing novel biology to identifying actionable genomic alterations. Cancer 122 20–33. (https://doi.org/10.1002/cncr.29706)
Bresler SC, Weiser DA, Huwe PJ, Park JH, Krytska K, Ryles H, Laudenslager M, Rappaport EF, Wood AC, McGrady PW, et al. 2014 ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell 26 682–694. (https://doi.org/10.1016/j.ccell.2014.09.019)
Brito JP & Hay ID 2017 Thyroid cancer: overdiagnosis of papillary carcinoma – who benefits? Nature Reviews Endocrinology 13 131–132. (https://doi.org/10.1038/nrendo.2016.224)
Brzezianska E, Pastuszak-Lewandoska D, Wojciechowska K, Migdalska-Sek M, Cyniak-Magierska A, Nawrot E & Lewinski A 2007 Investigation of V600E BRAF mutation in papillary thyroid carcinoma in the Polish population. Neuro Endocrinology Letters 28 351–359.
Cancer Genome Atlas Research Network 2014 Integrated genomic characterization of papillary thyroid carcinoma. Cell 159 676–690. (https://doi.org/10.1016/j.cell.2014.09.050)
Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C & Stuart JM 2013 The cancer genome atlas pan-cancer analysis project. Nature Genetics 45 1113–1120. (https://doi.org/10.1038/ng.2764)
Cancer Genome Atlas Research Network 2017 Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32 185.e113–203.e113. (https://doi.org/10.1016/j.ccell.2017.07.007)
Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, Schrock A, Campbell B, Shlien A, Chmielecki J, et al. 2017 Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Medicine 9 34. (https://doi.org/10.1186/s13073-017-0424-2)
Ciampi R, Mian C, Fugazzola L, Cosci B, Romei C, Barollo S, Cirello V, Bottici V, Marconcini G, Rosa PM, et al. 2013 Evidence of a low prevalence of RAS mutations in a large medullary thyroid cancer series. Thyroid 23 50–57. (https://doi.org/10.1089/thy.2012.0207)
Ciampi R, Romei C, Pieruzzi L, Tacito A, Molinaro E, Agate L, Bottici V, Casella F, Ugolini C, Materazzi G, et al. 2017 Classical point mutations of RET, BRAF and RAS oncogenes are not shared in papillary and medullary thyroid cancer occurring simultaneously in the same gland. Journal of Endocrinological Investigation 40 55–62. (https://doi.org/10.1007/s40618-016-0526-5)
Crona J, Taieb D & Pacak K 2017 New perspectives on pheochromocytoma and paraganglioma: toward a molecular classification. Endocrine Reviews 38 489–515. (https://doi.org/10.1210/er.2017-00062)
Cunningham JL, Diaz de Stahl T, Sjoblom T, Westin G, Dumanski JP & Janson ET 2011 Common pathogenetic mechanism involving human chromosome 18 in familial and sporadic ileal carcinoid tumors. Genes, Chromosomes and Cancer 50 82–94. (https://doi.org/10.1002/gcc.20834)
Dahia PL 2014 Pheochromocytoma and paraganglioma pathogenesis: learning from genetic heterogeneity. Nature Reviews Cancer 14 108–119. (https://doi.org/10.1038/nrc3648)
De Preter K, Mestdagh P, Vermeulen J, Zeka F, Naranjo A, Bray I, Castel V, Chen C, Drozynska E, Eggert A, et al. 2011 miRNA expression profiling enables risk stratification in archived and fresh neuroblastoma tumor samples. Clinical Cancer Research 17 7684–7692. (https://doi.org/10.1158/1078-0432.CCR-11-0610)
de Reynies A, Assie G, Rickman DS, Tissier F, Groussin L, Rene-Corail F, Dousset B, Bertagna X, Clauser E & Bertherat J 2009 Gene expression profiling reveals a new classification of adrenocortical tumors and identifies molecular predictors of malignancy and survival. Journal of Clinical Oncology 27 1108–1115. (https://doi.org/10.1200/JCO.2008.18.5678)
Esapa CT, Johnson SJ, Kendall-Taylor P, Lennard TW & Harris PE 1999 Prevalence of Ras mutations in thyroid neoplasia. Clinical Endocrinology 50 529–535. (https://doi.org/10.1046/j.1365-2265.1999.00704.x)
Fagin JA & Wells SA Jr 2016 Biologic and clinical perspectives on thyroid cancer. New England Journal of Medicine 375 1054–1067. (https://doi.org/10.1056/NEJMra1501993)
Fardin P, Barla A, Mosci S, Rosasco L, Verri A, Versteeg R, Caron HN, Molenaar JJ, Ora I, Eva A, et al. 2010 A biology-driven approach identifies the hypoxia gene signature as a predictor of the outcome of neuroblastoma patients. Molecular Cancer 9 185. (https://doi.org/10.1186/1476-4598-9-185)
Farrell WE 2014 Epigenetics of pituitary tumours: an update. Current Opinion in Endocrinology, Diabetes and Obesity 21 299–305. (https://doi.org/10.1097/MED.0000000000000078)
Favier J, Amar L & Gimenez-Roqueplo AP 2015 Paraganglioma and phaeochromocytoma: from genetics to personalized medicine. Nature Reviews Endocrinology 11 101–111. (https://doi.org/10.1038/nrendo.2014.188)
Feng H, Shuda M, Chang Y & Moore PS 2008 Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 319 1096–1100. (https://doi.org/10.1126/science.1152586)
Fishbein L, Leshchiner I, Walter V, Danilova L, Robertson AG, Johnson AR, Lichtenberg TM, Murray BA, Ghayee HK, Else T, et al. 2017 Comprehensive molecular characterization of pheochromocytoma and paraganglioma. Cancer Cell 31 181–193. (https://doi.org/10.1016/j.ccell.2017.01.001)
Francis JM, Kiezun A, Ramos AH, Serra S, Pedamallu CS, Qian ZR, Banck MS, Kanwar R, Kulkarni AA, Karpathakis A, et al. 2013 Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nature Genetics 45 1483–1486. (https://doi.org/10.1038/ng.2821)
George J, Lim JS, Jang SJ, Cun Y, Ozretic L, Kong G, Leenders F, Lu X, Fernandez-Cuesta L, Bosco G, et al. 2015 Comprehensive genomic profiles of small cell lung cancer. Nature 524 47–53. (https://doi.org/10.1038/nature14664)
Gibbons DL, Byers LA & Kurie JM 2014 Smoking, p53 mutation, and lung cancer. Molecular Cancer Research 12 3–13. (https://doi.org/10.1158/1541-7786.MCR-13-0539)
Giordano TJ 2011 The argument for mitotic rate-based grading for the prognostication of adrenocortical carcinoma. American Journal of Surgical Pathology 35 471–473. (https://doi.org/10.1097/PAS.0b013e31820bcf21)
Giordano TJ 2014 The cancer genome atlas research network: a sight to behold. Endocrine Pathology 25 362–365. (https://doi.org/10.1007/s12022-014-9345-4)
Giordano TJ 2018 Genomic hallmarks of thyroid neoplasia. Annual Review of Pathology 13 141–162. (https://doi.org/10.1146/annurev-pathol-121808-102139)
Giordano TJ, Kuick R, Else T, Gauger PG, Vinco M, Bauersfeld J, Sanders D, Thomas DG, Doherty G & Hammer G 2009 Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clinical Cancer Research 15 668–676. (https://doi.org/10.1158/1078-0432.CCR-08-1067)
Goh G, Walradt T, Markarov V, Blom A, Riaz N, Doumani R, Stafstrom K, Moshiri A, Yelistratova L, Levinsohn J, et al. 2016 Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 7 3403–3415. (https://doi.org/10.18632/oncotarget.6494)
Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA & Kurzrock R 2017 Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Molecular Cancer Therapeutics 16 2598–2608. (https://doi.org/10.1158/1535-7163.MCT-17-0386)
Goodwin S, McPherson JD & McCombie WR 2016 Coming of age: ten years of next-generation sequencing technologies. Nature Reviews Genetics 17 333–351. (https://doi.org/10.1038/nrg.2016.49)
Gortz B, Roth J, Krahenmann A, de Krijger RR, Muletta-Feurer S, Rutimann K, Saremaslani P, Speel EJ, Heitz PU & Komminoth P 1999 Mutations and allelic deletions of the MEN1 gene are associated with a subset of sporadic endocrine pancreatic and neuroendocrine tumors and not restricted to foregut neoplasms. American Journal of Pathology 154 429–436. (https://doi.org/10.1016/S0002-9440(10)65289-3)
Grubbs EG, Ng PK, Bui J, Busaidy NL, Chen K, Lee JE, Lu X, Lu H, Meric-Bernstam F, Mills GB, et al. 2015 RET fusion as a novel driver of medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 100 788–793. (https://doi.org/10.1210/jc.2014-4153)
Gu Y, Zhou X, Hu F, Yu Y, Xie T, Huang Y, Zhao X & Zhang X 2016 Differential DNA methylome profiling of nonfunctioning pituitary adenomas suggesting tumour invasion is correlated with cell adhesion. Journal of Neurooncology 129 23–31. (https://doi.org/10.1007/s11060-016-2139-4)
Gupta N, Dasyam AK, Carty SE, Nikiforova MN, Ohori NP, Armstrong M, Yip L, LeBeau SO, McCoy KL, Coyne C, et al. 2013 RAS mutations in thyroid FNA specimens are highly predictive of predominantly low-risk follicular-pattern cancers. Journal of Clinical Endocrinology and Metabolism 98 E914–E922. (https://doi.org/10.1210/jc.2012-3396)
Hanahan D & Weinberg RA 2011 Hallmarks of cancer: the next generation. Cell 144 646–674. (https://doi.org/10.1016/j.cell.2011.02.013)
Harms PW, Vats P, Verhaegen ME, Robinson DR, Wu YM, Dhanasekaran SM, Palanisamy N, Siddiqui J, Cao X, Su F, et al. 2015 The distinctive mutational spectra of polyomavirus-negative Merkel cell carcinoma. Cancer Research 75 3720–3727. (https://doi.org/10.1158/0008-5472.CAN-15-0702)
Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca A, Rizvi NA, Hirsch FR, Selvaggi G, Szustakowski JD, et al. 2018 Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 33 853.e4–861.e4. (https://doi.org/10.1016/j.ccell.2018.04.001)
Hiltzik D, Carlson DL, Tuttle RM, Chuai S, Ishill N, Shaha A, Shah JP, Singh B & Ghossein RA 2006 Poorly differentiated thyroid carcinomas defined on the basis of mitosis and necrosis: a clinicopathologic study of 58 patients. Cancer 106 1286–1295. (https://doi.org/10.1002/cncr.21739)
Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, Shen R, Taylor AM, Cherniack AD, Thorsson V, et al. 2018 Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173 291.e296–304.e296. (https://doi.org/10.1016/j.cell.2018.03.022)
Janoueix-Lerosey I, Schleiermacher G, Michels E, Mosseri V, Ribeiro A, Lequin D, Vermeulen J, Couturier J, Peuchmaur M, Valent A, et al. 2009 Overall genomic pattern is a predictor of outcome in neuroblastoma. Journal of Clinical Oncology 27 1026–1033. (https://doi.org/10.1200/JCO.2008.16.0630)
Jennings J & Hudson TJ 2013 Reflections on the founding of the International Cancer Genome Consortium. Clinical Chemistry 59 18–21. (https://doi.org/10.1373/clinchem.2012.184713)
Ji JH, Oh YL, Hong M, Yun JW, Lee HW, Kim D, Ji Y, Kim DH, Park WY, Shin HT, et al. 2015 Identification of driving ALK fusion genes and genomic landscape of medullary thyroid cancer. PLoS Genetics 11 e1005467. (https://doi.org/10.1371/journal.pgen.1005467)
Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, et al. 2011 DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331 1199–1203. (https://doi.org/10.1126/science.1200609)
Jochmanova I, Zhuang Z & Pacak K 2015 Pheochromocytoma: gasping for air. Hormones and Cancer 6 191–205. (https://doi.org/10.1007/s12672-015-0231-4)
Jung SH, Kim MS, Jung CK, Park HC, Kim SY, Liu J, Bae JS, Lee SH, Kim TM, Lee SH, et al. 2016 Mutational burdens and evolutionary ages of thyroid follicular adenoma are comparable to those of follicular carcinoma. Oncotarget 7 69638–69648. (https://doi.org/10.18632/oncotarget.11922)
Karpathakis A, Dibra H, Pipinikas C, Feber A, Morris T, Francis J, Oukrif D, Mandair D, Pericleous M, Mohmaduvesh M, et al. 2016 Prognostic impact of novel molecular subtypes of small intestinal neuroendocrine tumor. Clinical Cancer Research 22 250–258. (https://doi.org/10.1158/1078-0432.CCR-15-0373)
Kim HS, Lee JH, Nam SJ, Ock CY, Moon JW, Yoo CW, Lee GK & Han JY 2018a Association of PD-L1 expression with tumor-infiltrating immune cells and mutation burden in high-grade neuroendocrine carcinoma of the lung. Journal of Thoracic Oncology 13 636–648. (https://doi.org/10.1016/j.jtho.2018.01.008)
Kim M, Jeon MJ, Oh HS, Park S, Kim TY, Shong YK, Kim WB, Kim K, Kim WG & Song DE 2018b BRAF and RAS mutational status in noninvasive follicular thyroid neoplasm with papillary-like nuclear features and invasive subtype of encapsulated follicular variant of papillary thyroid carcinoma in Korea. Thyroid 28 504–510. (https://doi.org/10.1089/thy.2017.0382)
Kitahara CM & Sosa JA 2016 The changing incidence of thyroid cancer. Nature Reviews Endocrinology 12 646–653. (https://doi.org/10.1038/nrendo.2016.110)
Kober P, Boresowicz J, Rusetska N, Maksymowicz M, Goryca K, Kunicki J, Bonicki W, Siedlecki JA & Bujko M 2018 DNA methylation profiling in nonfunctioning pituitary adenomas. Molecular and Cellular Endocrinology [epub]. (https://doi.org/10.1016/j.mce.2018.01.020)
Koperek O, Kornauth C, Capper D, Berghoff AS, Asari R, Niederle B, von Deimling A, Birner P & Preusser M 2012 Immunohistochemical detection of the BRAF V600E-mutated protein in papillary thyroid carcinoma. American Journal of Surgical Pathology 36 844–850. (https://doi.org/10.1097/PAS.0b013e318246b527)
Kulke MH, Freed E, Chiang DY, Philips J, Zahrieh D, Glickman JN & Shivdasani RA 2008 High-resolution analysis of genetic alterations in small bowel carcinoid tumors reveals areas of recurrent amplification and loss. Genes, Chromosomes and Cancer 47 591–603. (https://doi.org/10.1002/gcc.20561)
Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, Dogan S, Ricarte-Filho JC, Krishnamoorthy GP, Xu B, et al. 2016 Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. Journal of Clinical Investigation 126 1052–1066. (https://doi.org/10.1172/JCI85271)
Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al. 2001 Initial sequencing and analysis of the human genome. Nature 409 860–921. (https://doi.org/10.1038/35057062)
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al. 2015 PD-1 blockade in tumors with mismatch-repair deficiency. New England Journal of Medicine 372 2509–2520. (https://doi.org/10.1056/NEJMoa1500596)
Levy SE & Myers RM 2016 Advancements in next-generation sequencing. Annual Review of Genomics and Human Genetics 17 95–115. (https://doi.org/10.1146/annurev-genom-083115-022413)
Llombart B, Requena C & Cruz J 2017 Update on Merkel cell carcinoma: epidemiology, etiopathogenesis, clinical features, diagnosis, and staging. Actas Dermo-Sifiliográficas 108 108–119. (https://doi.org/10.1016/j.ad.2016.07.022)
Lloyd RV, Osamura RY, Kloppel G & Rosai J 2017 WHO Classification of Tumors of Endocrine Organs. Lyon, France: IARC Press.
Ma ZY, Song ZJ, Chen JH, Wang YF, Li SQ, Zhou LF, Mao Y, Li YM, Hu RG, Zhang ZY, et al. 2015 Recurrent gain-of-function USP8 mutations in Cushing's disease. Cell Research 25 306–317. (https://doi.org/10.1038/cr.2015.20)
Mafficini A & Scarpa A 2018 Genomic landscape of pancreatic neuroendocrine tumours: the International Cancer Genome Consortium. Journal of Endocrinology 236 R161–R167. (https://doi.org/10.1530/JOE-17-0560)
McCain J 2006 The cancer genome atlas: new weapon in old war? Biotechnology Healthcare 3 46B–51B.
Medici M, Kwong N, Angell TE, Marqusee E, Kim MI, Frates MC, Benson CB, Cibas ES, Barletta JA, Krane JF, et al. 2015 The variable phenotype and low-risk nature of RAS-positive thyroid nodules. BMC Medicine 13 184. (https://doi.org/10.1186/s12916-015-0419-z)
Metzker ML 2010 Sequencing technologies – the next generation. Nature Reviews Genetics 11 31–46. (https://doi.org/10.1038/nrg2626)
Molenaar JJ, Koster J, Zwijnenburg DA, van Sluis P, Valentijn LJ, van der Ploeg I, Hamdi M, van Nes J, Westerman BA, van Arkel J, et al. 2012 Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes. Nature 483 589–593. (https://doi.org/10.1038/nature10910)
Morimoto R, Satoh F, Murakami O, Suzuki T, Abe T, Tanemoto M, Abe M, Uruno A, Ishidoya S, Arai Y, et al. 2008 Immunohistochemistry of a proliferation marker Ki67/MIB1 in adrenocortical carcinomas: Ki67/MIB1 labeling index is a predictor for recurrence of adrenocortical carcinomas. Endocrine Journal 55 49–55. (https://doi.org/10.1507/endocrj.K07-079)
Nicolson NG, Murtha TD, Dong W, Paulsson JO, Choi J, Barbieri AL, Brown TC, Kunstman JW, Larsson C, Prasad ML, et al. 2018 Comprehensive genetic analysis of follicular thyroid carcinoma predicts prognosis independent of histology. Journal of Clinical Endocrinology and Metabolism [epub]. (https://doi.org/10.1210/jc.2018-00277)
Nikiforov YE, Seethala RR, Tallini G, Baloch ZW, Basolo F, Thompson LD, Barletta JA, Wenig BM, Al Ghuzlan A, Kakudo K, et al. 2016 Nomenclature revision for encapsulated follicular variant of papillary thyroid carcinoma: a paradigm shift to reduce overtreatment of indolent tumors. JAMA Oncology 2 1023–1029. (https://doi.org/10.1001/jamaoncol.2016.0386)
Paulson KG, Lemos BD, Feng B, Jaimes N, Penas PF, Bi X, Maher E, Cohen L, Leonard JH, Granter SR, et al. 2009 Array-CGH reveals recurrent genomic changes in Merkel cell carcinoma including amplification of L-Myc. Journal of Investigative Dermatology 129 1547–1555. (https://doi.org/10.1038/jid.2008.365)
Paulson VA, Shivdasani P, Angell TE, Cibas ES, Krane JF, Lindeman NI, Alexander EK & Barletta JA 2017 Noninvasive follicular thyroid neoplasm with papillary-like nuclear features accounts for more than half of ‘carcinomas’ harboring RAS mutations. Thyroid 27 506–511. (https://doi.org/10.1089/thy.2016.0583)
Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, et al. 2013 The genetic landscape of high-risk neuroblastoma. Nature Genetics 45 279–284. (https://doi.org/10.1038/ng.2529)
Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F, Meitinger T, Mizuno-Yamasaki E, Kawaguchi K, Saeki Y, et al. 2015 Mutations in the deubiquitinase gene USP8 cause Cushing's disease. Nature Genetics 47 31–38. (https://doi.org/10.1038/ng.3166)
Ronchi CL, Peverelli E, Herterich S, Weigand I, Mantovani G, Schwarzmayr T, Sbiera S, Allolio B, Honegger J, Appenzeller S, et al. 2016 Landscape of somatic mutations in sporadic GH-secreting pituitary adenomas. European Journal of Endocrinology 174 363–372. (https://doi.org/10.1530/EJE-15-1064)
Rossi G, Bertero L, Marchio C & Papotti M 2018 Molecular alterations of neuroendocrine tumours of the lung. Histopathology 72 142–152. (https://doi.org/10.1111/his.13394)
Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, et al. 2018 Oncogenic signaling pathways in the cancer genome atlas. Cell 173 321.e310–337.e310. (https://doi.org/10.1016/j.cell.2018.03.035)
Sausen M, Leary RJ, Jones S, Wu J, Reynolds CP, Liu X, Blackford A, Parmigiani G, Diaz LA Jr, Papadopoulos N, et al. 2013 Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nature Genetics 45 12–17. (https://doi.org/10.1038/ng.2493)
Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, et al. 2017 Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543 65–71. (https://doi.org/10.1038/nature21063)
Schleiermacher G, Janoueix-Lerosey I, Ribeiro A, Klijanienko J, Couturier J, Pierron G, Mosseri V, Valent A, Auger N, Plantaz D, et al. 2010 Accumulation of segmental alterations determines progression in neuroblastoma. Journal of Clinical Oncology 28 3122–3130. (https://doi.org/10.1200/JCO.2009.26.7955)
Schwab M, Ellison J, Busch M, Rosenau W, Varmus HE & Bishop JM 1984 Enhanced expression of the human gene N-myc consequent to amplification of DNA may contribute to malignant progression of neuroblastoma. PNAS 81 4940–4944. (https://doi.org/10.1073/pnas.81.15.4940)
Simbolo M, Mafficini A, Sikora KO, Fassan M, Barbi S, Corbo V, Mastracci L, Rusev B, Grillo F, Vicentini C, et al. 2017 Lung neuroendocrine tumours: deep sequencing of the four World Health Organization histotypes reveals chromatin-remodelling genes as major players and a prognostic role for TERT, RB1, MEN1 and KMT2D. Journal of Pathology 241 488–500. (https://doi.org/10.1002/path.4853)
Steuer CE & Ramalingam SS 2018 Tumor mutation burden: leading immunotherapy to the era of precision medicine? Journal of Clinical Oncology 36 631–632. (https://doi.org/10.1200/JCO.2017.76.8770)
Stratton MR, Campbell PJ & Futreal PA 2009 The cancer genome. Nature 458 719–724. (https://doi.org/10.1038/nature07943)
Stricker TP, Morales La Madrid A, Chlenski A, Guerrero L, Salwen HR, Gosiengfiao Y, Perlman EJ, Furman W, Bahrami A, Shohet JM, et al. 2014 Validation of a prognostic multi-gene signature in high-risk neuroblastoma using the high throughput digital NanoString nCounter system. Molecular Oncology 8 669–678. (https://doi.org/10.1016/j.molonc.2014.01.010)
Swarts DR, Ramaekers FC & Speel EJ 2012 Molecular and cellular biology of neuroendocrine lung tumors: evidence for separate biological entities. Biochimica et Biophysica Acta 1826 255–271. (https://doi.org/10.1016/j.bbcan.2012.05.001)
Theodros D, Patel M, Ruzevick J, Lim M & Bettegowda C 2015 Pituitary adenomas: historical perspective, surgical management and future directions. CNS Oncology 4 411–429. (https://doi.org/10.2217/cns.15.21)
Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, Schernthaner-Reiter MH, Szarek E, Leal LF, Caberg JH, et al. 2014 Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. New England Journal of Medicine 371 2363–2374. (https://doi.org/10.1056/NEJMoa1408028)
Tufton N, Roncaroli F, Hadjidemetriou I, Dang MN, Denes J, Guasti L, Thom M, Powell M, Baldeweg SE, Fersht N, et al. 2017 Pituitary carcinoma in a patient with an SDHB mutation. Endocrine Pathology 28 320–325. (https://doi.org/10.1007/s12022-017-9474-7)
Turchini J, Cheung VKY, Tischler AS, De Krijger RR & Gill AJ 2018 Pathology and genetics of phaeochromocytoma and paraganglioma. Histopathology 72 97–105. (https://doi.org/10.1111/his.13402)
Uraki S, Ariyasu H, Doi A, Furuta H, Nishi M, Sugano K, Inoshita N, Nakao N, Yamada S & Akamizu T 2017 Atypical pituitary adenoma with MEN1 somatic mutation associated with abnormalities of DNA mismatch repair genes; MLH1 germline mutation and MSH6 somatic mutation. Endocrine Journal 64 895–906. (https://doi.org/10.1507/endocrj.EJ17-0036)
Valenciaga A, Grubbs EG, Porter K, Wakely PE Jr, Williams MD, Cote GJ, Vasko VV, Saji M & Ringel MD 2017 Reduced retinoblastoma protein expression is associated with decreased patient survival in medullary thyroid cancer. Thyroid 27 1523–1533. (https://doi.org/10.1089/thy.2017.0113)
Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, Smith HO, Yandell M, Evans CA, Holt RA, et al. 2001 The sequence of the human genome. Science 291 1304–1351. (https://doi.org/10.1126/science.1058040)
Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr & Kinzler KW 2013 Cancer genome landscapes. Science 339 1546–1558. (https://doi.org/10.1126/science.1235122)
Volante M, Collini P, Nikiforov YE, Sakamoto A, Kakudo K, Katoh R, Lloyd RV, LiVolsi VA, Papotti M, Sobrinho-Simoes M, et al. 2007 Poorly differentiated thyroid carcinoma: the Turin proposal for the use of uniform diagnostic criteria and an algorithmic diagnostic approach. American Journal of Surgical Pathology 31 1256–1264. (https://doi.org/10.1097/PAS.0b013e3180309e6a)
Volante M, Bussolati G & Papotti M 2016 The story of poorly differentiated thyroid carcinoma: from Langhans' description to the Turin proposal via Juan Rosai. Seminars in Diagnostic Pathology 33 277–283. (https://doi.org/10.1053/j.semdp.2016.05.007)
Wang Z, Jensen MA & Zenklusen JC 2016 A practical guide to the cancer genome atlas (TCGA). Methods in Molecular Biology 1418 111–141.
Weiss LM 1984 Comparative histologic study of 43 metastasizing and nonmetastasizing adrenocortical tumors. American Journal of Surgical Pathology 8 163–169. (https://doi.org/10.1097/00000478-198403000-00001)
Weiss LM, Medeiros LJ & Vickery AL Jr 1989 Pathologic features of prognostic significance in adrenocortical carcinoma. American Journal of Surgical Pathology 13 202–206. (https://doi.org/10.1097/00000478-198903000-00004)
Westermark UK, Wilhelm M, Frenzel A & Henriksson MA 2011 The MYCN oncogene and differentiation in neuroblastoma. Seminars in Cancer Biology 21 256–266. (https://doi.org/10.1016/j.semcancer.2011.08.001)
Wong HL, Yang KC, Shen Y, Zhao EY, Loree JM, Kennecke HF, Kalloger SE, Karasinska JM, Lim HJ, Mungall AJ, et al. 2017 Molecular characterisation of metastatic pancreatic neuroendocrine tumours (PNETs) using whole genome and transcriptome sequencing. Cold Spring Harbor Molecular Case Studies 4 a002329. (https://doi.org/10.1101/mcs.a002329)
Yachida S, Vakiani E, White CM, Zhong Y, Saunders T, Morgan R, de Wilde RF, Maitra A, Hicks J, Demarzo AM, et al. 2012 Small cell and large cell neuroendocrine carcinomas of the pancreas are genetically similar and distinct from well-differentiated pancreatic neuroendocrine tumors. American Journal of Surgical Pathology 36 173–184. (https://doi.org/10.1097/PAS.0b013e3182417d36)
Yang Z, Zhang T & Gao H 2016 Genetic aspects of pituitary carcinoma: a systematic review. Medicine 95 e5268. (https://doi.org/10.1097/MD.0000000000005268)
Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE, Abdalla EK, Fleming JB, Vauthey JN, Rashid A, et al. 2008 One hundred years after ‘carcinoid’: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. Journal of Clinical Oncology 26 3063–3072. (https://doi.org/10.1200/JCO.2007.15.4377)
Yarchoan M, Hopkins A & Jaffee EM 2017 Tumor mutational burden and response rate to PD-1 inhibition. New England Journal of Medicine 377 2500–2501. (https://doi.org/10.1056/NEJMc1713444)
Zhang J, Baran J, Cros A, Guberman JM, Haider S, Hsu J, Liang Y, Rivkin E, Wang J, Whitty B, et al. 2011 International cancer genome consortium data portal – a one-stop shop for cancer genomics data. Database 2011 bar026. (https://doi.org/10.1093/database/bar048)
Zhao L, Dias-Santagata D, Sadow PM & Faquin WC 2017 Cytological, molecular, and clinical features of noninvasive follicular thyroid neoplasm with papillary-like nuclear features versus invasive forms of follicular variant of papillary thyroid carcinoma. Cancer Cytopathology 125 323–331. (https://doi.org/10.1002/cncy.21839)
Zheng S, Cherniack AD, Dewal N, Moffitt RA, Danilova L, Murray BA, Lerario AM, Else T, Knijnenburg TA, Ciriello G, et al. 2016 Comprehensive pan-genomic characterization of adrenocortical carcinoma. Cancer Cell 29 723–736. (https://doi.org/10.1016/j.ccell.2016.04.002)
Zhu Z, Gandhi M, Nikiforova MN, Fischer AH & Nikiforov YE 2003 Molecular profile and clinical-pathologic features of the follicular variant of papillary thyroid carcinoma. An unusually high prevalence of ras mutations. American Journal of Clinical Pathology 120 71–77. (https://doi.org/10.1309/ND8D9LAJTRCTG6QD)