Skip to main content
The Journal of Clinical Endocrinology and Metabolism logoLink to The Journal of Clinical Endocrinology and Metabolism
. 2025 Feb 5;110(10):e3283–e3302. doi: 10.1210/clinem/dgaf075

Overlapping Presentations and Diverse Genetic Defects Characterize Neuroendocrine Neoplasms in a Mexican Cohort

Laura C Hernández-Ramírez 1,, Claudia Ramírez-Rentería 2, Rosa G Rebollar-Vega 3, Julia M Zuarth-Vázquez 4, Mariana Torres-Morán 5, Alexa L Franco-Álvarez 6, Andrea I Manzo-Osnaya 7, Edgar Martell-Jiménez 8, Maribel Rodríguez-Torres 9, Wellbert E Hernández-Núñez 10, Alfredo A Reza-Albarrán 11, Armando Gamboa-Domínguez 12, Etual Espinosa-Cárdenas 13, Ernesto Sosa-Eroza 14, Irma Hernández-García 15, Baldomero González-Virla 16, Alex F Hernández-Martínez 17, Manuel García-Sáenz 18, Aldo Eguiluz-Melendez 19, Moisés Mercado 20, Guadalupe Vargas-Ortega 21, Lourdes Balcázar-Hernández 22
PMCID: PMC12448596  PMID: 39908201

Abstract

Context

Genetic tests are part of the routine clinical approach to syndromic and nonsyndromic phenotypes of neuroendocrine neoplasms (NENs). Current data on phenotype–genotype associations in NENs, however, do not accurately represent all populations.

Objective

To describe the frequency, inventory, and clinical associations of germline defects associated with multiple types of NENs in a Mexican cohort.

Methods

Blood DNA from Mexican adults with NENs was analyzed with a 53-gene next-generation sequencing panel developed ad hoc (n = 90) or Sanger sequencing (n = 2). Single nucleotide variants, indels, and structural variants were identified, classified, and subjected to orthogonal confirmation. When possible, tumor samples and blood DNA from additional family members were tested using Sanger sequencing.

Results

Ninety-two probands (70.7% women, 51.5% sporadic) were included; 16 carried pathogenic or likely pathogenic (P/LP) variants and were significantly younger at disease onset than the rest (29.6 ± 10.7 vs 40 [21.5-51.5] years, P = .0384). Likely driving variants were identified in three-quarters of Von Hippel Lindau syndrome cases, one-third of multiple endocrine neoplasia (MEN) type 1, one-quarter of early-onset acromegaly/gigantism, and individual cases of Cushing's disease, MEN2A, and medullary thyroid carcinoma. One patient with clinical MEN1 associated with an SDHA variant and 1 with a pituitary tumor and neurofibromatosis type 1 were also identified. Probands with familial disease were more likely to carry P/LP variants than sporadic cases (26.7 vs 8.5%, P = .0282).

Conclusion

P/LP variants were identified in 17.4% of individuals with NENs. Our research provides a view of the landscape of NEN drivers in a population not previously characterized.

Keywords: familial isolated pituitary adenoma, multiple endocrine neoplasia, neuroendocrine neoplasm, next-generation sequencing, paraganglioma/pheochromocytoma, pituitary neuroendocrine tumor


Neuroendocrine neoplasms (NENs) comprise a heterogeneous group of lesions derived from cells with the capacity for producing hormones (1-4). These tumors may arise from various tissues, presenting either individually or as part of syndromes of multiple endocrine neoplasia (MEN) (5). Their clinical behavior is quite variable, spanning from indolence to hormone oversecretion, compromise of surrounding structures, frank aggressiveness, and malignancy (6-8). Although traditionally considered rare, the frequency of NENs has recently increased, due to improved clinical awareness and novel diagnostic methods (9-11).

NENs entail a very strong heritable component: around 40% of pheochromocytomas and paragangliomas (PPGLs), 10% to 20% of gastrointestinal NENs (GI-NENs), pancreatic NENs (panNENs), bronchopulmonary, and thymic NENs, 16% to 25% of cases of medullary thyroid carcinoma (MTC), up to 15% of parathyroid NENs, and 5% to 10% of pituitary neuroendocrine tumors (PitNETs) are due to germline defects (12-19). The genetic drivers involved include MEN–associated genes (CDKN1B, MAX, MEN1, PRKAR1A, and RET), defects leading to inherited isolated NENs (loss of function [LOF] variants in AIP and SDHx or GPR101 gene amplification), alterations causing syndromes of both inherited NENs and cancer (DICER1, TP53, and VHL), and genes causing phakomatoses where NENs are often a manifestation (NF1, PTEN, TSC1, and TSC2) (18, 20-27). In addition, somatic genetic changes, such as loss of heterozygosity (LOH) and hotspot variants (affecting BRAF, GNAS, SF3B1, USP8, and USP48 in PitNETs) might serve as biomarkers for particular tumor phenotypes, while others (RET in MTC) represent potential therapeutic targets (28-31).

In the last decade, the widespread use of powerful tools for genetic analyses has resulted in a dramatic increase in the known genetic causes of NENs. These approaches have also yielded instances of unexpected genetic diagnoses for particular phenotypes that include NENs, uncovering a great overlap in their clinical presentation (32-36). Genetic tests are key for early diagnosis, tailored clinical management, and genetic counseling in patients with NENs (23, 37-39). Yet, published data on the frequency and type of genetic disease drivers do not accurately represent all human populations. In particular, genetic tests are not widely available in Mexico and are almost nonexistent in its public hospitals, which serve the majority of the country's ∼129 million population. Precision medicine strategies require population-specific information, but Mexicans, one of the most genetically diverse human groups, are poorly represented in international genetic databases (40, 41). Therefore, we aimed to explore the type and frequency of germline tumor drivers in a prospective cohort of Mexican individuals with NENs and to define the phenotypes and clinical outcomes associated with specific genetic defects.

Materials and Methods

Patients and Samples

Eligible participants were adults (≥18 years, with no maximum age limit) with a new or previous diagnosis of NENs under medical care at either of 2 participating reference hospitals in Mexico City: Hospital de Especialidades de Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Individuals with previously established genetic diagnosis were not excluded. The research protocol was approved by the internal review boards of both institutions (protocol IDs R-2022-785-011 and 4090, respectively; ClinicalTrials.gov: NCT06523582). All participants provided written informed consent and a peripheral blood sample at recruitment. Relatives of probands with positive genetic analyses and 3 apparently healthy controls from the same population were also included under informed consent.

Three groups of diagnoses were considered for recruitment: (1) isolated NENs with sporadic presentation (bronchopulmonary NENs, GI-NENs, or panNENs, MTC, PPGLs, PitNETs, and primary hyperparathyroidism [PHPT]); (2) familial isolated NENs (familial isolated pituitary adenoma [FIPA], familial MTC [FMTC], familial PPGLs, familial PHPT, familial gastrointestinal stromal tumors [GISTs], and X-linked acrogigantism); and (3) clinical syndromes encompassing NENs and other manifestations with familial or sporadic presentation (Carney complex, Carney–Stratakis syndrome, Carney triad, Cowden syndrome, DICER1 syndrome, Li–Fraumeni syndrome, Lynch syndrome, MEN-like syndrome [MEN-like], MEN1, MEN2A and 2B, neurofibromatosis type 1 [NF1], Pacak–Zhuang syndrome, PPGL and pituitary adenoma syndrome, tuberous sclerosis complex, and von Hippel–Lindau syndrome [VHL]). Definitions for clinical diagnoses and family history within each group are detailed in supplemental material (42). Exclusion criteria were age <18 years, unavailable clinical data, unwillingness to provide a blood sample, and inability to provide informed consent.

Recruitment started in August 2022 and, as of September 2024, 198 individuals (169 probands and 29 additional family members) had been included. Ninety-four individuals, including 2 patients with previously established genetic diagnoses via Sanger sequencing, were included in the present analysis. Clinical data were obtained directly from the participants and/or their clinical records. Fresh frozen tumor samples were obtained from patients that underwent surgery during the study. When available, formalin-fixed and paraffin-embedded (FFPE) samples were retrieved from the pathology archives. DNA was extracted from blood using the DNA Isolation Kit for Mammalian Blood (Roche 11667327001) plus RNase (Roche 11119915001) treatment. For DNA extraction from tissues, samples were first mechanically disrupted with magnetic beads (fresh frozen tissues) or deparaffinized (FFPE samples) with HistoChoice (Merck H2779) and then processed with the Maga Zorb DNA Mini-Prep kit (Promega MB1004).

Targeted Next-Generation Sequencing

A panel for capture-based next-generation sequencing (NGS) was designed using the Twist Biosciences platform, covering all exons, 5′ and 3′ untranslated regions (UTRs), and exon–intron junctions (50 bp outside of exons) of the following genes: AIP (11q13.2, reference transcript: NM_003977.4), ATRX (Xq21.1, NM_000489.6), BRAF (7q34, NM_004333.6), CABLES1 (18q11.2, NM_001100619.3), CDKN1A (6p21.2, NM_000389.5), CDKN1B (12p13.1, NM_004064.5), CDKN2B (9p21.3, NM_004936.4), CDKN2C (1p32.3, NM_078626.3), CDC73 (1q31.2, NM_024529.5), DAXX (6p21.32, NM_001141969.2), DICER1 (14q32.13, NM_177438.3), DLST (14q24.3, NM_001933.5), DNMT3A (2p23.3, NM_022552.5), EGLN1 (1q42.2, NM_022051.3), EPAS1 (2p21, NM_001430.5), FH (1q43, NM_000143.4), GNAS (20q13.32, NM_000516.7), GOT2 (16q21, NM_002080.4), GPR101 (Xq26.3, NM_054021.2), H3-3A (1q42.12, NM_002107.7), KIF1B (1p36.22, NM_001365951.3), KIT (4q12, NM_000222.3), MAFA (8q24.3, NM_201589.4), MAX (14q23.3, NM_002382.5), MDH2 (7q11.23, NM_005918.4), MEN1 (11q13.1, NM_130799.3), MET (7q31.2, NM_000245.4), MLH1 (3p22.2, NM_000249.4), MSH2 (2p21, NM_000251.3), MSH6 (2p16.3, NM_000179.3), MERTK (2q13, NM_006343.3), NF1 (17q11.2, NM_001042492.3), PDGFRA (4q12, NM_006206.6), PIK3CA (3q26.32, NM_006218.4), PMS2 (7p22.1, NM_000535.7), PRKAR1A (17q24.2, NM_002734.5), PTEN (10q23.31, NM_000314.8), RASD1 (17p11.2, NM_016084.5), RET (10q11.21, NM_020975.6), SDHA (5p15.33, NM_004168.4), SDHAF2 (11q12.2, NM_017841.4), SDHB (1p36.13, NM_003000.3), SDHC (1q23.3, NM_003001.5), SDHD (11q23.1, NM_003002.4), SLC25A11 (17p13.2, NM_003562.5), SUCLG2 (3p14.1, NM_003848.4), TMEM127 (2q11.2, NM_017849.4), TP53 (17p13.1, NM_000546.6), TSC1 (9q34.13, NM_000368.5), TSC2 (16p13.3, NM_000548.5), USP8 (15q21.2, NM_005154.5), USP48 (1p36.12, NM_032236.8), and VHL (3p25.3, NM_000551.4). The final design (Twist Custom Panel TE-95598353) includes 2023 probes, spans 208 790 bp, and covers 99.5% of the 209 381 bp of the intended target region.

For library preparation, ∼400 bp fragments were obtained by mechanical fragmentation of 200 ng blood DNA in a Covaris S220 sonicator. End repair and dA-tailing, adapter ligation, and amplification, as well as hybridization and postcapture amplification of 8 × 250 ng sample pools were carried out by means of the Twist Biosciences Mechanical Fragmentation and Universal Adapter and Target Enrichment Standard Hybridization protocols. DNA fragment size analyses after sonication, indexing, and hybridization were performed by means of the DNA 1000 or DNA high sensitivity kits (Agilent 5067-1504 and 5067-4626), as appropriate, in a 2100 Bioanalyzer (Agilent). DNA at these steps was quantified with the Qubit dsDNA HS or dsDNA BR Assay Kits (Invitrogen Q32854 and Q32853), as appropriate. Sequencing of 8 pM library pools with 1.25% PhiX Control v3 (Illumina FC-110-3001) was carried out using the MiSeq Reagent Kit v2 (300-cycles, Illumina MS-102-2002) in a MiSeq (Illumina) instrument to obtain paired-end 150 bp reads with an aimed 300× sequencing depth.

Sequencing Data Analyses

Quality was verified using FastQC and reads were then aligned to the GRCh38/hg38 human genome using Burrows–Wheeler aligner-maximum exact matches (43, 44). Polymerase chain reaction (PCR) duplicates were marked with MarkDuplicates and FreeBayes was used for variant calling (45, 46). Copy number variants were searched for with ExomeDepth (47). The Franklin (Genoox) online platform was used to select medium and high confidence nonsynonymous variants in the regions of interest, with sequencing depth ≥20 (48). Using public online database browsers, variants with frequency <0.1% in The Genome Aggregation Database v.4.0, the indigenous Mexican subset of samples from the Mexico City Population Database, and All of Us were then selected (41, 49, 50). For all samples, a manual screen of NGS raw data for the variants of interest was performed, using the Integrative Genomics Viewer 2.3.72 platform (51). Variants of interest were searched for in ClinVar and their reported classification (according to the criteria of the American College of Medical Genetics and Genomics and Association for Molecular Pathology, ACMG/AMP) was noted (52, 53). For variants with conflicting classification or not listed in ClinVar, functional effects were predicted using the in silico tools linked to the Varsome and Franklin online platforms and available clinical and functional data were retrieved using the Mastermind (Genomenon) online platform (54, 55). Based on those data, ACMG/AMP categories were assigned. All variants are reported according to the Human Genome Variation Society nomenclature (56).

Sanger Sequencing and Droplet Digital PCR

All pathogenic and likely pathogenic (P/LP) variants and selected variants of uncertain significance (VUS) identified by NGS were subjected to orthogonal confirmation, using Sanger sequencing for single nucleotide variants (SNVs) and indels or droplet digital (dd) PCR for structural variants (SVs). The same methods were used to analyze the germline variant loci in FFPE or fresh frozen tumor samples. Sanger sequencing was also used to confirm variants of interest in 2 patients with a previous diagnostic test, as well as to investigate BRAF, USP8, and USP48 hotspot variants in 6 FFPE corticotropinomas. The regions of interest were amplified by end-point PCR, using specific primers and GoTaq Green Master Mix (Promega M7122). Amplicons were column-purified (Wizard SV Gel and PCR Clean-Up System, Promega A9281) and subjected to unidirectional sequencing (BigDye Terminator 3.1 Cycle Sequencing Kit, Applied Biosystems 4337456) in a 3500xL Genetic Analyzer (Applied Biosystems). Sequences were analyzed using the Geneious Prime v.2024.0.5 (Biomatters, Ltd.) software.

Reactions for ddPCR were prepared using 6-carboxyfluorescein-labeled assays targeting the regions of interest, a 2′-chloro-7′-phenyl-1,4-dichloro-6-carboxyfluorescein-labeled RPP30 assay (Applied Biosystems 4403326) as an internal control, HindIII restriction enzyme (New England Biolabs, NEB, R3104), and ddPCR SuperMix for Probes (Bio-Rad 1863024). Droplet generation and reading were done in a QX200 Droplet Digital PCR System (Bio-Rad). Results were analyzed with the Quanta Soft software 1.7.4.0917 (Bio-Rad). Primers and assays for end-point PCR, Sanger sequencing, and ddPCR are listed in Table S1 (42). A commercial predesigned assay (Applied Biosystems TaqMan 6-carboxyfluorescein-labeled Hs02125257_cn) was used for ddPCR for VHL exon 3.

Performance of Sequencing Platform

Peripheral blood DNA samples of 92 probands and 3 controls were sequenced using the custom-designed NGS panel, achieving a median sequencing depth of 243.5x (223-259.9) for the 53 genes analyzed (Fig. S1 (42)). Only DAXX yielded a low mean depth of sequencing, at 60.3 ± 8.1x, probably due to the presence of low complexity regions. Due to quality concerns, samples from 2 probands were not considered for further analyses. All SNVs, indels and SVs of interest passed orthogonal confirmation, except for 2 defects in PMS2 and SDHC, which were considered artifactual. In the first gene, exon 13 variant c.2182_2184delinsG, p.T728Afs*7 was found in heterozygosity in 2 probands and passed all selection filters. This region displays homology with the pseudogene PMS2CL, and PCR using primers in specific regions of PMS2 followed by Sanger sequencing ruled out the variant (57). In a different individual, testing of a deletion in exon 5 of SDHC by ddPCR rendered dubious results (4.6 vs 5.2 copies in a control), also likely due to a pseudogene (58).

Statistical Analyses

Analyses were carried out using Prism 10.2.3 (GraphPad Software). For numerical variables, data distribution was analyzed using the Shapiro–Wilk test. Parametric data are presented as mean ± SD and nonparametric data are presented as median and interquartile range. Age at disease onset and at diagnosis were compared among groups using the Mann–Whitney test. The frequency of P/LP variants among sporadic and familial cases was compared using the Fisher exact test. The odds ratio is presented with 95% confidence interval in brackets, and was calculated using the Baptista–Pike method. P < .05 was considered statistically significant.

Results

Clinical Features

We present data for 92 probands (70.7% women, Fig. 1A), 90 studied via NGS panel and 2 with previous Sanger sequencing–based genetic diagnoses. The mean age at recruitment was 48.8 ± 16.9 years and the median ages at disease onset and diagnosis were 37 (21-49.5) and 40 (23.3-50.8) years, respectively. The clinical presentation was classified into 15 different phenotypes, being the most common MEN1, MEN-like, and early-onset PitNETs without growth hormone (GH) secretion, with 16, 13, and 13 individuals, respectively (Fig. 1B). Most individuals (51.1%, n = 47) had no NEN-related family history. Among nonsporadic cases, the most common familial phenotype was cancer predisposition, in one-fifth of probands (Fig. 1C).

Figure 1.

Figure 1.

Clinical characteristics of the study population and general description of genetic findings. (A) Sex distribution of the probands (n = 92). (B) Categories of clinical presentation of the study probands (n = 92). The categories were determined according to the clinical features observed at diagnosis and during follow-up. (C) Family history in probands (n = 92). Patients with no relevant family history were classified as sporadic and the rest were grouped into 10 categories, according to the phenotypes described in the family members. (D) All variants (n = 303) identified in the study, according to their predicted effect. (E) ACMG/AMP categories for all variants identified in the study (n = 303). (F) Comparison of the age at disease onset and diagnosis between individuals with a detected pathogenic or likely pathogenic variant (P/LP) and the rest of probands (Rest). The horizontal bar represents median, and the error bars depict interquartile range. CS, Cushing's syndrome; del, deletion; dup, duplication; FIPA, familial isolated pituitary adenoma; FMTC, familial medullary thyroid carcinoma; FPPGLs, familial pheochromocytomas/paragangliomas; gig/EO acro, gigantism and early onset acromegaly; ins, insertion; ins/del, insertion/deletion; LB, likely benign; LFL, Li–Fraumeni-like; LP, likely pathogenic; MEN-like, syndrome similar to multiple endocrine neoplasia; MEN1, multiple endocrine neoplasia type 1; MEN2A, multiple endocrine neoplasia type 2A; MTC, medullary thyroid carcinoma; M GI-NENs, multiple gastrointestinal neuroendocrine neoplasms; M panNENs, multiple pancreatic neuroendocrine neoplasms; NF1, neurofibromatosis type 1; non-GH EO PitNETs, early onset PitNETs not causing GH excess; panNEN, pancreatic neuroendocrine neoplasm; PitNET, pituitary neuroendocrine tumor; PPGL, pheochromocytoma/paraganglioma; VHL, Von Hippel Lindau syndrome; VUS, variants of uncertain significance.

Results of Genetic Analyses

In total, 303 genetic variants were identified, almost half of which (n = 143) were missense (Fig. 1D). One-hundred and eighty-one variants were benign, 67 were likely benign (LB), 35 were VUS, 14 were P/LP, and 6 were not classified (Fig. 1E). All variants were found at the germline level, except for USP8 p.S718P, which was a somatic change in 1 corticotropinoma. The 248 different benign and LB variants identified in the study are listed in Table S2 (42).

The P/LP variants included 12 SNVs or indels and 2 SVs, affecting 8 genes (AIP, MEN1, NF1, RET, SDHA, SDHD, VHL, and USP8) and were found as heterozygous changes in 16 cases (17.4% of those analyzed, Table 1). Two variants, RET p.C618R and VHL p.L89P, were detected in 2 cases each, while the rest affected individual probands. MEN1 was the most frequently affected gene, with 5 P/LP variants in 5 different individuals with MEN1 (3 sporadic and 2 familial), including 3 novel changes. Two variants per gene were found for VHL and AIP, while the rest of genes displayed single variants. Probands carrying P/LP variants had a significantly younger age at disease onset (29.6 ± 10.7 years) than the rest (40 [21.5-51.5] years, P = .0384). These individuals were also diagnosed slightly earlier (32.6 ± 11.3 years), although not at a significantly younger age than other probands (40.5 [24-54.3] years, P = .1250, Fig. 1F).

Table 1.

Clinical associations of pathogenic and LP variants in the study cohort

Varianta Frequency in general population (%) ACMG/AMP classification Reported clinical and experimental data Case Clinical presentation Family history and targeted screening
gnomAD MCPS IMX All of Us ClinVar (accession) This study
AIP (NM_003977.4)
c.713G>A, p.C238Y
(rs267606569) germline
0.0001 n/a n/a LP (VCV000041197.9) LP Missense variant in exon 5. Results in an unstable protein with loss of multiple molecular interactions. Cosegregation proven in 1 family with FIPA (83) 1 Male, AR: 22 y. Gigantism: AO 14 y (accelerated growth, prognathism, headaches, DKA), Ad 18 y (macroadenoma); TSS (improvement, active tumor remnant), LAN 120 mg monthly (partial control), RT to tumor remnant planned. Lost to follow-up Apparently sporadic. Family n/a
c.910C>T, p.R304* (rs104894195) germline 0.0014 n/a 0.0006 Pathogenic (VCV000004888.33) P Nonsense variant in exon 6. Most frequent germline AIP variant causing FIPA. Results in a truncated and unstable protein with loss of multiple molecular interactions (83) 2 Male, AR: 50 y. Gigantism: AO 15 y (accelerated growth), Ad 18 y (macroadenoma); TSS (18 y, improvement with tumor remnant, then recurrence), BEC (improvement, then tumor growth), and RT (remission with hypopituitarism, empty sella) FIPA: 1 paternal first cousin (female) with young-onset acromegaly. F and 1 sister are apparently healthy variant carriers, 1 sister is not a carrier
MEN1 (NM_130799.3)
c.(?_-23-27)_(445 + 65_?)del germline n/a n/a n/a n/a P Large deletion spanning intron 1, exon 2, and intron 2. Not previously reported; predicted to cause LOF (truncated protein) 3 Female, AR: 21 y. MEN1: 1. Two panNENs, AO 19 y (recurrent severe hypoglycemia), Ad 19 y (17, 21 mm tumors); subtotal pancreatectomy (21 y, well-differentiated NET grade 1, 2 mitoses/10 HPF, Ki-67 2%, insulin + <1% cells; remission)
2. PHPT (AO/Ad 21 y, targeted biochemical screening); 3½ gland parathyroidectomy (21 y, remission, ddPCR: 1 copy of MEN1 exon 2 in fresh frozen tumor). Underwent uneventful pregnancy (22 y) and delivered a healthy baby
Apparently sporadic. M and brother are not variant carriers. Paternal family n/a
c.496C>T, p.Q166* germline n/a n/a n/a Pathogenic (VCV000618208.9) P Nonsense variant in exon 3. Predicted to cause LOF (truncated protein). Previously reported germline variant in individuals with MEN1 (84, 85) and as a somatic change in 1 porocarcinoma (86) and 1 somatotropinoma (87) 4 Male, AR: 42 y. MEN1:
1. PHPT, AO 25 y (recurrent nephrolithiasis), Ad 37 y
2. Meningioma (AO/Ad 30 y), surgically resected
3. PanNEN (AO/Ad 36 y); subtotal pancreatectomy
Familial MEN1: multiple cousins of F had PHPT. PGF died of prostate cancer, brother with recurrent UTIs. M and 2 sisters are not variant carriers. Paternal family n/a
c.660G>A, p.W220* (rs886039414) germline n/a n/a n/a Pathogenic (VCV000265235.14) P Nonsense variant in exon 4. Predicted to cause LOF (truncated protein). Previously reported in multiple families with MEN1 (88-90), including 1 with 2 thymic NEN cases (63) and 1 with a case of malignant melanoma (91), as well as in 1 panNEN (92) 5b Male, AR: 43 y. MEN1:
1. Pancreatic head neuroendocrine carcinoma, AO/Ad 34 y (jaundice, 47 × 47 mm tumor); Whipple procedure (34 y, well-differentiated neuroendocrine pancreatic carcinoma, synaptophysin and CgA +, no metastases, Ki-67 20%, positive surgical margins; metastases 2 y later), LAN 90, then 120 mg monthly (since 40 y, improvement, progression 3 y later), 177Lu-DOTATATE (44 y, 7 cycles, stable stage IV).
2. PHPT (AO/Ad: 34 y, targeted screening); 3½ gland parathyroidectomy + thymectomy.
3. Thymic NEN: AO/Ad: 34 y (incidental finding during follow-up, regrowth in image study 10 y after resection)
4. PitNET, AO/Ad: 34 y (targeted screening, 2 mm). Developed CS (41 y), pending localization studies
Familial MEN1: affected M, younger brother (PHPT), 1 maternal uncle (PHPT and panNEN), 1 maternal aunt (PHPT and thymic NEN). Three maternal uncles with recurrent nephrolithiasis. One apparently healthy daughter, affected younger brother, and maternal aunt are variant carriers. M is obligate carrier. One brother and 1 sister tested negative for variant
c.936_938del, p.Y313del germline n/a n/a n/a n/a LP In-frame deletion in exon 7. Not previously reported; predicted to cause LOF 6 Female, AR: 38. MEN1:
1. Gastric NEN (AO/Ad: 36 y, GERD, polyp); endoscopic resection (gastric lesion: well-differentiated neuroendocrine tumor, grade 1 WHO, CKAE1-AE3 +, enolase +, synaptophysin + 100%, chromogranin + 100%, Ki-67 1%)
2. Multiple panNENs (AO/Ad: 37 y, incidental finding in CT scan, n = 7: 27, 16, 16, 6, 5, 11, and 6 mm); subtotal pancreatectomy
3. PHPT (AO/Ad: 37 y, targeted screening); 3½ gland parathyroidectomy (diffuse hyperplasia in 3 glands). LOH at variant locus in fresh frozen parathyroid tissue
Apparently sporadic. M and sister are not variant carriers. Paternal family n/a
c.984_985del, p.H328Qfs*38 germline n/a n/a n/a n/a LP Frameshift variant in exon 7. Not previously reported; predicted to cause LOF (truncated protein). Another variant with the same effect on the protein (c.984_990del, p.H328Qfs*38) was reported in 1 case of MEN1 (93). Other variants in the same residue have been associated with MEN1 (94) 7 Male, AR: 52 y. MEN1:
1. PHPT: AO 36 y (nephrolithiasis)/Ad: 40 y; resection of 1 and 2 glands in 2 surgeries, then resection of remnant gland with implant on forearm (1 gland with adenoma, 1 with hyperplasia)
2. PanNEN, AO/Ad: 40 y (recurrent severe hypoglycemia); subtotal pancreatectomy (multiple panNENs, the largest of 16 mm, glucagon +)
3. Prolactinoma, AO/Ad: 42 y (targeted screening, microadenoma); CBG
Apparently sporadic
NF1 (NM_001042492.3):
c.147C>A, p.Y49* germline n/a n/a n/a Pathogenic (VCV002114679.2) P Nonsense variant in exon 2. Predicted to cause LOF (truncated protein). Reported as a germline change in patients with NF1 (95-97) and at the somatic level in 1 case of ovarian cancer (98) and 1 of synchronous endometrial and ovarian cancer (99) 8 Male, AR: 54. NF1:
1. Multiple cutaneous neurofibromas
2. Café-au-lait spots
3. NF-PitNET (AO/Ad 54 y, headaches, hemianopsia, memory impairment and seizures, 16 × 15 × 15 mm tumor); TSS (54 y, PitNET, 0 mitoses/2 mm2, weak/hypogranular PAS, chromogranin +, ACTH, GH, and prolactin –, Ki-67 < 1%). No LOH at NF1 variant locus. Improved symptoms, tumor remnant not compromising vision or gland function
Familial NF1: café-au-lait spots in F, 8/10 siblings, and daughter. One nephew died (28 y) from complications of epilepsy. Affected daughter and 2 apparently healthy grandchildren are variant carriers.c History of endogamy. From small community with multiple affected individuals
RET (NM_020975.6)
c.1852T>C, p.C618R (rs76262710) germline 0.0001 n/a n/a Pathogenic (VCV000013905.89) P Missense variant in exon 10. Results in a protein with constitutive oncogenic activity and altered expression in vitro (100, 101). Reported as a germline change in multiple families with MEN2 and FMTC, proven cosegregation (102-105) and in cases of Hirschsprung disease (106). ATA-MOD (39) 9 Male, AR: 64 y. MEN2A:
1. PHEO, AO/Ad 33 y; surgical resection
2. MTC, AO/Ad 61 y; TT and selective neck dissection. Currently in remission
Familial MEN2A: M, sister and 1 son with MTC, 1 niece with PHEO. Affected son and niece, and apparently healthy daughter are variant carriers; 3 sisters and 1 brother are not carriersc
10 Female, AR 35 y. MTC, AO 30 y (palpable neck mass)/Ad 35 y; TT and selective neck dissection (MTC with lymphatic, perineural, and muscle invasion, calcitonin +++, CEA +++, Tg –, Ki67 +++), radical neck dissection (persistent disease), RT FMTC: F and 6 half-siblings with unspecified disease who required TT, 1 paternal first cousin (female) with MTC
SDHA (NM_004168.4)
c.2T>A, p.M1? (rs750380279) germline 0.0001 0.0016 0.0020 Pathogenic (VCV000582115.13) P Start loss variant in exon 1; predicted to cause LOF. Previously reported at the germline level in 1 patient with carotid body PGL (107). Other variants with the same effect have been reported in PPGLs (108) 11 Male, AR 59 y. MEN1:
1. Acromegaly, AO 36 y (acral growth), Ad 51 y (13 mm PitNET); LAN 120 mg monthly, TSS (active remnant), LAN 120 mg monthly + CBG, RT.
2. PHPT, AO/Ad 52 y (nephrolithiasis); right inferior parathyroidectomy + TT
3. Thyroid nodule, AO/Ad 52 y (finding during studies for PHPT); follicular adenoma
Apparently sporadic. F had nephrolithiasis; brother had liver neoplasm (primary?)
SDHD (NM_003002.4)
c.320T>G, p.L107R (rs876658477) germline 0.0001 0.0022 n/a Conflicting classifications of pathogenicity: pathogenic, LP, VUS (VCV000230274.27) LP Missense variant in exon 4. In silico analyses: deleterious effect. Germline defect in 1 case of carotid body PGL (109). Different missense variant in this residue (p.L107P) reported in familial PPGLs, with proven cosegregation (110) 12 Female, AR 40 y. Bilateral carotid body PPGLs:
AO 30 y (palpable neck masses, paroxystic high blood pressure), Ad 42 y (right 47 × 36 × 57 mm, left: 27 × 12 × 31 mm); RT, OCT LAR. Tumor growth in last evaluation
Family with cancer predisposition. Maternal uncle with tongue cancer, paternal aunt with Bc
VHL (NM_000551.4)
c.266T>C, p.L89P (rs5030807) germline n/a n/a n/a Pathogenic/LP (VCV000182979.12) P Missense variant in exon 1. Predicted to disrupt protein function. Previously reported as germline change in multiple individuals and families with VHL (111-123), including 1 family from Mexico (124). Reported in ccRCC as a germline (125) and somatic variant (126-129), and in CNS HB (130) 13 Male, AR 42. VHL:
1. Vermian cerebellar HB (AO/Ad 41 y)
2. Pulmonary nodule (27 × 33 × 24 mm)
3. Uncinate process panNEN (by PET-CT, no biopsy)
4. Bilateral renal cysts
5. Left ccRCC
Familial VHL:
F died of bladder cancer; M with renal cyst
14b Female, AR 36 y. VHL:
1. Pancreatic adenocarcinoma, AO/Ad 22 y; Whipple procedure, chemo, RT
2. Left ccRCC, AO/Ad 31 y; partial left nephrectomy
3. Right ovary serose cystadenoma, AO/Ad 30 y (incidental finding during follow-up); resection of right adnexa
4. Right retinal hemangioma, AO/Ad 30 y (targeted screening)
5. Signet ring cell adenocarcinoma of gastro-jejunal anastomosis, AO 34 y (weight loss), Ad 35 y (24 × 45 mm); partial gastrectomy + open Roux-en-Y gastric bypass, chemo. Deceased
Familial VHL: MGM died of gastric cancer, affected M, maternal aunt, 4 maternal first cousins, brother, and sister are variant carriers; 1 unaffected maternal aunt and 1 sister are apparently unaffected variant carriers; 2 sisters and 1 brother are not carriersc
c.(?_464-50)_(*50_?)del germline n/a n/a n/a n/a P Large deletion spanning intron 2 and exon 3 up to 3′-UTR. Not previously reported; predicted to cause LOF (truncated protein) 15 Male, AR 38 y. VHL:
  1. Retinal angioma (AO/Ad 28 y)

  2. PanNEN (AO/Ad 29 y); subtotal pancreatectomy

  3. Renal cysts (AO/Ad 29 y); under surveillance

  4. Multiple intracranial HBs (AO/Ad 29 y)

  5. Pulmonary nodules (AO/Ad 29 y); under surveillance

Familial VHL: 1 sister, F, and paternal uncle with VHL; paternal aunt with renal tumors; paternal aunt with suspected kidney disease; PGM died of renal cancer; 4 brothers of PGM and 4 children of 1 of them with probable VHL. From small community with multiple affected individuals
USP8 (NM_005154.5)
c.2152T>C, p.S718P somatic 0.0001 0.0000 0.0000 Pathogenic (VCV000161992.1) P Missense variant in exon 14 hotspot. One of the most common USP8 variants in CD. Leads to loss of interaction with 14-3-3, increased USP8 cleavage/DUB activity, reduced EGFR degradation, and enhanced ACTH suppression with pasireotide in vitro (131) 16b Female, AR 21 y. CD: AO 15 y (CS), Ad 21 y, (PitNET 8.4 × 4.3 × 3.6 mm). Treated with KTCZ (21 y), then TSS (21 y, densely granulated corticotropinoma, ACTH +, TSH, FSH, LH, prolactin, and GH –, CK1/AE3 +, Ki-67 < 2%; active tumor remnant); currently on KTCZ n/a (somatic variant)

Abbreviations: AD, age at diagnosis; AO, age at onset; AR, age at recruitment; ATA-MOD, American Thyroid Association, moderate risk of MTC; BC, breast cancer; BEC, bromocriptine; CBG, cabergoline; ccRCC, clear cell renal cell carcinoma; CD, Cushing's disease; CEA, carcinoembryonic antigen; CNS, central nervous system; CS, Cushing's syndrome; CT, computed tomography; ddPCR, droplet digital polymerase chain reaction; DKA, diabetic ketoacidosis; DUB, deubiquitinase; F, father; FMTC, familial MTC; HB, hemangioblastoma; HPF, high-power fields; KTCZ, ketoconazole; LAN, lanreotide; LOF, loss-of-function; LP, likely pathogenic; M, mother; MEN, multiple endocrine neoplasia; MGM, maternal grandmother; MTC, medullary thyroid carcinoma; n/a, not available; NEN, neuroendocrine neoplasm; NET, neuroendocrine tumor; OCT LAR, octreotide long-acting release; P, pathogenic; panNEN, pancreatic NEN; PET, positron emission tomography; PGF, paternal grandfather; PGL, paraganglioma; PGM, paternal grandmother; PHEO, pheochromocytoma; PHPT, primary hyperparathyroidism; pitNET, pituitary neuroendocrine tumor; PPGL, pheochromocytoma and paraganglioma; RT, radiotherapy; Tg, thyroglobulin; TT, total thyroidectomy; TSS, transsphenoidal surgery; UTIs, urinary tract infections; UTR, untranslated region; VHL, Von Hippel-Lindau syndrome; VUS, variant of uncertain significance.

a dbSNP ID in parentheses, if available.

b Variant detection was done with Sanger sequencing.

c Family screening in a different laboratory.

The 35 VUS were heterozygous, occurred in single cases, and affected 19 different genes (Table 2). Previous publications proposed a pathogenic effect for 4 of these variants. AIP p.R106C was detected in a patient with neuroendocrine gastric carcinoma and multiple gastric type 1 NENs (Case 17). This variant was previously reported in a patient with a prolactinoma and family history of PHPT (59). Also affecting AIP, p.V291_L292del was found in an individual with early-onset acromegaly in the setting of FIPA (Case 18), but the additional affected family member has not been tested. This variant was originally identified in a sporadic case of gigantism in a Mexican patient and is predicted to disrupt the packaging of the C-terminal α-helix of AIP (60). Regarding these cases, it must be noted that PitNETs account for the only known clinical association of germline LOF AIP defects. CDKN1B p.I119T was identified in a patient with sporadic gigantism (Case 19). Previous reports of this defect in the literature include 1 case of acromegaly within a FIPA family and 1 case of pediatric Cushing's disease (CD); CDKN1B LOF is the cause of MEN4 (26). Finally, the MEN1 p.M558del variant, included in ClinVar, was found in 1 case of sporadic MEN1 (Case 28). The region affected by this in-frame deletion is required for multiple molecular interactions and contains variants with a known pathogenic effect. Functional studies and testing of additional family members are required to accurately categorize these genetic changes.

Table 2.

VUS identified via NGS panel in the study cohort

Varianta Frequency in general population (%) ACMG/AMP classification Case Clinical presentation (family history)
gnomAD MCPS IMX All of Us ClinVar (accession) This manuscript
AIP (NM_003977.4)
c.316C>T, p.R106C (rs369414668) 0.0075 0.0027 0.0043 VUS (VCV000485059.10) VUS 17 Neuroendocrine gastric carcinoma and multiple gastric type 1 NENs (sporadic)
c.872_877del, p.V291_L292del n/a n/a 0.0004 n/a VUS 18 Early-onset acromegaly (FIPA)
CDKN1B (NM_004064.5)
c.356T>C, p.I119T
(rs142833529)
0.0706 0.0016 0.0722 Conflicting interpretations of pathogenicity: VUS, benign, LB (VCV000307664.28) VUS 19 Gigantism (sporadic)
c.482C>T, p.S161F (rs373917399) 0.0007 0.0547 0.0014 Conflicting interpretations of pathogenicity: VUS, LB (VCV000582241.11) VUS 20b Insulinoma (familial cancer predisposition)
FH (NM_000143.4)
c.1431_1433dup, p.K477dup (rs367543046) 0.1915 n/a 0.1373 Conflicting interpretations of pathogenicity: pathogenic, LP, VUS (VCV000042095.81) VUS 21 Acromegaly (suspected FIPA)
GNAS (NM_000516.7)
Upstream from NM_000516.7; c.*42 + 12949T>C, p.? in NM_016592.5 n/a 0.0060 0.0004 VUS (VCV002355759.2) VUS 22 Bilateral carotid body PGL (Li–Fraumeni-like)
Upstream from NM_000516.7; c.*42 + 13032C>T, p.? in NM_016592.5 (rs1354669859) 0.0006 0.0026 0.0008 n/a VUS 23 MEN-like: ccRCC, left adrenal tumor, 2 panNENs, interhemispheric lipoma, multiple colorectal adenomas (familial cancer predisposition)
Upstream from NM_000516.7;c.*42 + 13400G>A, p.? in NM_016592.5 (rs777234998) 0.0013 n/a 0.0004 n/a VUS 24 MEN-like: right testicular non-seminomatous germ cell tumor and left adrenal PHEO (sporadic)
KIF1B (NM_001365951.3)
c.1898G>A, p.R633H (rs1650295604) 0.0001 0.0016 n/a VUS (VCV001779575.2) VUS 25 NF-PitNET, PHPT, PTC, uterine fibroids and benign left breast tumor (MEN1, sporadic)
MAFA (NM_201589.4):
c.109G>A, p.E37K (rs529585367) 0.0006 n/a 0.0018 n/a VUS 26 Gigantism (sporadic)
MDH2 (NM_005918.4)
c.236-4C>G, p.? (rs1554586397) 0.0002 n/a 0.0004 n/a VUS 27 Multiple gastric NENs (sporadic)
MEN1 (NM_130799.3)
c.1673_1675del, p.M558del n/a n/a n/a VUS (VCV001777737.2) VUS 28 MEN1: insulinomatosis, GH and prolactin-secreting PitNET, multiple parathyroid adenomas (familial MEN1)
MERTK (NM_006343.3)
c.1396_1397delinsGA, p.R466E (rs386649210) n/a n/a n/a VUS (VCV001414697.5) VUS 20b See above
c.2590G>A, p.V864I (rs557004700) 0.0025 0.0358 0.0043 VUS (VCV000893540.6) VUS 29 MEN-like: PHPT and left aldosterone-producing ACA (sporadic)
c.2674C>G, p.P892A 0.0001 0.0011 n/a n/a VUS 30b MEN1: NF-PitNET and PHPT (sporadic)
c.2689A>G, p.I897V (rs775526399) 0.0001 0.0077 0.0004 n/a VUS 31 MEN-like: meningioma and panNEN (sporadic)
c.2947G>A, p.D983N (rs781065147) 0.0128 0.0463 0.0137 VUS (VCV001504094.6) VUS 32c MEN-like: NF-PitNET and multiple lipomas (sporadic)
MET (NM_000245.4)
c.*23C>G, p.? (rs980940721) 0.0008 n/a 0.0055 n/a VUS 33 MTC (sporadic)
MSH6 (NM_000179.3)
c.30C>A, p.F10L (rs786201869) 0.0001 n/a n/a VUS (VCV000628563.10) VUS 32c See above
c.1601A>T, p.N534I (rs1572723598) n/a 0.0097 n/a VUS (VCV000819644.8) VUS 3d See Table 1
c.2331G>T, p.W777C (rs876660037) 0.0001 0.0021 n/a VUS (VCV001428821.6) VUS 34 Bilateral carotid body PGL (sporadic)
c.3059A>G, p.N1020S n/a 0.0005 n/a VUS (VCV001006155.7) VUS 32c See above
PDGFRA (NM_006206.6)
c.-13 + 5029G>C (rs546074770) n/a 0.0151 0.0002 n/a VUS 32c See above
c.1432_1436delinsCCCCA, p.S478_R479delinsPQ n/a n/a n/a n/a VUS 35 PHPT, PHEO (MEN2A, sporadic)
c.1631T>C, p.V544A (rs181854060) 0.0084 0.0011 0.0334 Conflicting interpretations of pathogenicity: LP, VUS, LB (VCV000039617.19) VUS 24 See above
c.2323 + 1243G>T (rs1052140725) 0.0013 n/a 0.0059 n/a VUS 26 See above
c.3221_3222delinsGC, p.D1074G n/a n/a n/a VUS (VCV000950280.5) VUS 36 Early-onset prolactinoma (sporadic)
PMS2 (NM_000535.7)
c.209A>G, p.D70G (rs1554305000) n/a 0.0003 0.0002 VUS (VCV000484322.15) VUS 37 Gastrinoma (sporadic)
c.1139T>G, p.V380G (rs748971523) 0.0002 0.0272 0.0008 VUS (VCV000630207.14) VUS 38 Early-onset acromegaly (sporadic)
PTEN (NM_000314.8)
c.-348G>C n/a n/a 0.0002 n/a VUS 30b See above
SDHA (NM_004168.4)
c.1396G>A, p.A466T (rs111387770) 0.2992 n/a n/a Conflicting interpretations of pathogenicity: VUS, benign, LB (VCV000472327.21) VUS 2d See Table 1
SDHC (NM_003001.5)
c.180-3C>T (rs1328389180) 0.0001 0.0027 0.0002 VUS (VCV001331275.7) VUS 36 See above
TMEM127 (NM_017849.4)
c.*542C>G, p.? 0.0047 n/a 0.0075 n/a VUS 24 See above
TP53 (NM_000546.6)
c.871A>G, p.K291E (rs1555525126) 0.0001 0.0162 0.0004 VUS (VCV000480951.17) VUS 39 Right carotid body PGL (Li–Fraumeni-like)
VHL (NM_000551.4)
c.65A>G, p.E22G (rs2125124570) 0.0001 0.0114 0.0004 VUS (VCV001403440.10) VUS 40 MTC (sporadic)

Abbreviations: ACA, adrenal cortex adenoma; ccRCC, clear cell renal cell carcinoma; FIPA, familial isoalted pituitary adenoma; LB, likely benign; LP, likely pathogenic; MEN, multiple endocrine neoplasia; MEN-like, syndrome similar to MEN; MTC, medullary thyroid carcinoma; n/a, not available; NEN, neuroendocrine neoplasm; NF-PitNET, non-functional PitNET; panNEN, pancreatic NEN; PGL, paraganglioma; PHEO, pheochromocytoma; PHPT, primary hyperparathyroidism; PitNET, pituitary neuroendocrine tumor; PTC, papillary thyroid carcinoma; VUS, variant of uncertain significance.

a dbSNP ID in parentheses, if available.

b Two coexisting VUS in different genes.

c Four coexisting VUS in 4 different genes.

d This patient carries a causative variant in a different gene.

Since the association of CABLES1 variants with CD has been proposed, but not confirmed, an ACMG/AMP category was not assigned to 6 variants identified in this gene (61). Out of them, 3 rare variants were detected in Case 25 (p.S17R), Case 27 (p.A155S), and in a patient with MEN2A and negative RET screening (p.R412W). The variant p.E178 K, which affects the ability of CABLES1 to reduce cell proliferation in vitro, was found in Case 11. This individual carried a different germline variant that could explain the phenotype (see “Genotype–Phenotype associations”). Such variants could represent disease modifiers, although further research is required to test this hypothesis.

Screening in Additional Family Members

For 6 cases with variants of interest, 21 additional family members were recruited for the study. Nineteen of them underwent targeted screening to determine their carrier status, using Sanger sequencing (n = 19) or ddPCR (n = 2). Five variant carriers were identified in 3 families, 3 carrying AIP variants (c.910C>T, p.R304* in 2 individuals and p.V291_L292del in 1), and 2 members of 1 family carrying MEN1 p.W220*. The AIP variant carriers are 77, 75, and 46 years old and are apparently unaffected. Biochemical and imaging testing was advised only for the youngest patient and remains pending. Both MEN1 variant carriers, aged 41 and 56 years at recruitment, underwent biochemical and imaging testing and were found to be affected with PHPT and PHPT and thymic NEN, respectively.

Genotype–Phenotype Associations

Fifteen out of the 16 patients with P/LP variants had clinical presentations previously associated with the affected genes. As expected, three-quarters of patients with clinical VHL (75%) carried pathogenic VHL variants (Table 3). In line with the high penetrance of this entity, all cases had positive family history. The only patient with clinical VHL and negative genetic testing has also a history of hereditary hemorrhagic telangiectasia, but no other relatives with VHL-associated features. A defect in a gene different than VHL (perhaps ELOC) might be the underlying cause of his phenotype (62).

Table 3.

Genetic findings in probands according to clinical categories

Clinical categories Genetic findingsa Total, no. (%)
P/LP, no. (%) VUS, no. (%) No VOI, no. (%)
Recruitment groups b
Sporadic isolated NENs 2 (6.9) 10 (34.5) 17 (58.6) 29 (100)
Familial isolated NENs 3 (11.5) 5 (19.2) 18 (69.2) 26 (100)
Familial or sporadic syndromic NENs 11 (29.7) 9 (24.3) 17 (45.9) 37 (100)
Total: 16 (17.4) 24 (26.1) 52 (56.5) 92 (100)
Clinical presentation in probands
Ectopic Cushing's syndrome 2 (100) 2 (100)
Gigantism or early onset acromegaly 2 (25) 4 (50) 2 (25) 8 (100)
MEN-like 5 (38.5) 8 (61.5) 13 (100)
MEN1 6 (37.5) 3 (18.8) 7 (43.8) 16 (100)
MEN2A 1 (33.3) 1 (33.3) 1 (33.3) 3 (100)
MTC 1 (12.5) 2 (25) 5 (62.5) 8 (100)
Multiple GI-NENs 1 (50) 1 (50) 2 (100)
Multiple panNENs 1 (100) 1 (100)
Multiple PPGLs 1 (10) 2 (20) 7 (70) 10 (100)
NF1 1 (100) 1 (100)
Early onset PitNETs not causing GH excess 1 (7.7) 1 (7.7) 11 (84.6) 13 (100)
PanNEN 2 (40) 3 (60) 5 (100)
PitNET 1 (25) 3 (75) 4 (100)
Single PPGL 1 (50) 1 (50) 2 (100)
VHL 3 (75) 1 (25) 4 (100)
Total: 16 (17.4) 24 (26.1) 52 (56.5) 92 (100)
Family history in probands
Sporadic 4 (8.5) 18 (38.3) 25 (53.2) 47 (100)
Cancer predisposition 1 (5) 2 (10) 17 (85) 20 (100)
FIPA 1 (16.7) 2 (33.3) 3 (50) 6 (100)
Familial MTC 1 (100) 1 (100)
Familial PPGLs 1 (100) 1 (100)
Li Fraumeni-like 2 (100) 2 (100)
MEN-like 5 (100) 5 (100)
MEN1 4 (80) 1 (20) 5 (100)
MEN2A 1 (100) 1 (100)
NF1 1 (100) 1 (100)
VHL 3 (100) 3 (100)
Total: 16 (17.4) 24 (26.1) 52 (56.5) 92 (100)

Abbreviations: FIPA, familial isolated pituitary adenoma; FPPGL, familial pheochromocytoma and paraganglioma; GI-NEN, gastrointestinal NEN; MEN, multiple endocrine neoplasia; MEN-like, syndrome similar to multiple endocrine neoplasm; MTC, medullary thyroid carcinoma; NEN, neuroendocrine neoplasm; NF1, neurofibromatosis type 1; P/LP, pathogenic/likely pathogenic; panNEN, pancreatic NEN; PPGL, pheochromocytoma and paraganglioma; VHL, Von Hippel-Lindau syndrome; VOI, variant of interest; VUS, variant of uncertain significance.

a Individuals with more than 1 type of variant were included in the most clinically relevant category.

b Clinical presentation in the family (according to available data) was used to classify cases as isolated or syndromic.

Regarding clinical MEN1, 5 out of 16 patients (31.3%) carried P/LP MEN1 variants, and 1 carried a novel VUS, accounting for 37.5% of patients with this phenotype if both P/LP and VUS are considered. All these patients developed panNENs and PHPT and all, except the 2 youngest ones, had also PitNETs. Interestingly, in Case 5, both the proband and another carrier of MEN1 p.W220* were diagnosed with thymic NEN, a rare occurrence that has been previously associated with this variant (63). Out of the 10 patients with clinical MEN1 and negative MEN1 screening, only 1 individual presented with the association of panNEN, PitNET, and PHPT, while the rest developed PitNETs (6 nonfunctioning PitNETs, 1 somatotropinoma, and 1 corticotropinoma) and PHPT, but no panNENs. One of these individuals (Case 11) carried a pathogenic SDHA variant with loss of the start codon. This patient presented with acromegaly and PHPT, a particular combination of components of MEN1 that usually tests negative for defects in MEN1-associated genes (64). To our knowledge, this is the first report of a patient with clinical MEN1 associated with this genetic defect. Two other MEN1-negative patients, Cases 25 and 27, carried VUS in KIF1B (p.R633H) and MERTK (p.P892A), respectively. These genes have not been associated with the genotype before.

P/LP AIP variants were identified in 2 out of 8 individuals with early-onset acromegaly or gigantism (25%). If 2 additional patients carrying a VUS of interest in AIP (p.V291_L292del) and CDKN1B (p.I119T) are considered, the frequency of variants of interest in this group rises to 50%. Out of these 4 cases, 2 presented in the setting of FIPA and 2 had sporadic gigantism. Among the 4 remaining cases, Case 38 (sporadic young-onset acromegaly) carried a PMS2 VUS and Case 26 (sporadic gigantism) had VUS in 2 genes not associated with his phenotype. The remaining 2 patients (sporadic gigantism and early-onset acromegaly with family history of cancer predisposition) had no variants of interest identified. In the subgroup of young onset PitNETs not secreting GH, only 1 out of 13 individuals (7.1%) had a clear tumor driver (somatic USP8 variant). One patient (Case 36, early-onset prolactinoma) carried 2 VUS in genes not previously associated with the phenotype; the rest had negative screening. None of the patients with isolated PitNETs with age at onset >30 years (n = 4) carried P/LP variants but Case 21 carries a VUS in FH, a gene not associated with this phenotype. Interestingly, a pathogenic NF1 variant was found in a single patient with NF1 who developed a PitNET, which is not a typical component of the syndrome

Among patients presenting with MEN2A (n = 3) or MTC (n = 8), 2 carried a pathogenic RET variant, 1 with familial MEN2A and 1 with FMTC. Among the 9 remaining subjects, 8 presented sporadically and 1 had MEN-like family history. Three of these individuals carried VUS in genes not previously associated with their phenotypes: Case 33 (sporadic MTC, MET 3′-UTR variant), Case 35 (sporadic MEN2A, PDGFRA p.S478_R479delinsPQ), and Case 40 (sporadic MTC, VHL p.E22G).

One out of 10 individuals with multiple PPGLs analyzed carried a pathogenic variant in SDHD (p.L107R). Two individuals carried VUS in GNAS and MSH6; these genes are not associated with the phenotype. None of the patients with single PPGLs carried P/LP variants, but 1 patient with a Li–Fraumeni-like family history carried a VUS in TP53 (p.K291E). Unfortunately, in this case no samples from tissues or additional family members were available for study.

Finally, no P/LP variants were identified for isolated single or multiple panNENs (n = 6), multiple GI-NENs (n = 2), MEN-like phenotypes (n = 13), or ectopic Cushing's syndrome (CS) (n = 2). VUS were found in 2 patients with GI-NENs (Cases 17 and 27), including the abovementioned AIP p.R106C, and an intronic MERTK variant. Also, 2 patients with panNENs had VUS. Case 37 (sporadic gastrinoma) carried PMS2 p.D70G and Case 20 carried 2 variants: MERTK p.R466E and CDKN1B p.S161F. Regarding the latter variant, a different missense change in the same residue (p.S161C) was reported as a germline defect in a recently published case of multiple panNENs, but there are no available functional or cosegregation data for this variant (65). Among individuals with MEN-like presentations, VUS were detected in 5 individuals: an intronic GNAS variant was identified in Case 23, Cases 29 and 31 carried MERTK variants, Case 24 had 1 PDGFRA and 1 TMEM127 VUS, and Case 32 carried variants in MERTK, MSH6 (2 variants), and PDGFRA. Additional validation is required to determine the role of the identified variants on this heterogeneous phenotype.

Patients with a familial presentation were significantly more likely to carry a P/LP variant, compared with sporadic cases (26.7 vs 8.5%, odds ratio 3.9 [1.2-11.7], P = .0282). P/LP variants were detected in all patients with familial VHL (n = 3), familial NF1 (n = 1), familial MEN2A (n = 1), and FMTC (n = 1), as well as 4 out of 5 cases of familial MEN1, 1 out of 6 cases of FIPA, and 1 out of 20 cases with cancer predisposition. No P/LP variants were found in cases with family history of MEN-like (n = 5), Li–Fraumeni-like (n = 2), and familial PPGLs (n = 1). Therefore, a positive family history for the specific phenotype present in the proband is a strong predictor of P/LP variants.

Discussion

We developed and validated a custom designed NGS panel that can be applied for the genetic diagnosis of multiple types of NENs. Three main characteristics distinguish this research platform from available routine NGS panels for the different categories of NENs included in the study. First, the selection of genes is designed to cover the most frequent, although not all, germline and somatic defects involved in these tumors. This design allowed for a “medium sized” panel, thus reducing sequencing costs and simplifying data analyses. Second, 2 genes of uncertain significance with a suggested, but not fully proven, role in the pathogenesis of corticotropinoma (CABLES1 and RASD1) were included; genes of uncertain significance are not typically part of routine panels (66). Third, cancer-related genes without a known implication in NENs are not covered, thus reducing the possibility of reportable secondary findings. We believe that this simplified strategy for genetic testing for multiple types of NENs will be useful for widespread routine use.

Variant classification in this population was challenging due to the finding of variants that are extremely rare in other human groups. Fortunately, during the course of the study, a large dataset of genetic variants in general Mexican population, MCPS, became available (41). Also, in the last years, the Hispanic/Latino subset of samples in the All of Us project has considerably grown (50). These data greatly eased our work, thus highlighting the importance of population-matched reference databases for variant interpretation. Our own dataset was also useful for variant classification, even though the cohort is still small. Remarkably, 4 out of 14 P/LP variants (3 in MEN1 and 1 in VHL) have not been reported before. As the number of participants grows, it will become clear whether these population-specific variants display a founder effect. Also, 11 variants not listed in ClinVar were classified as benign due to their high frequency in MCPS and/or Hispanic individuals from All of Us. Four other defects absent from ClinVar were classified as LB based on in silico prediction plus frequency ≥5% in our dataset. Finally, DICER1 c.4206+9_4206+21delinsT, reported in ClinVar as a VUS, was reclassified as LB due to the high allele frequency observed in our samples (18.3%, including 3 homozygous individuals).

Most of the clinical associations of the genetic defects identified in our study are well-known, and in most cases our findings align with previous studies. For instance, LOF MEN1 variants explain 90% of MEN1 cases with positive family history, but only one-third of those with sporadic presentation; therefore, these defects drive 70% to 90% of all MEN1 cases (67-70). We detected P/LP MEN1 variants in one-third of all probands with MEN1: 12.5% (1/8) of those with sporadic presentation and 50% (4/8) of those with positive family history. An additional sporadic MEN1 case was associated with an MEN1 VUS that requires further validation. To our knowledge, there are no previous reports of the frequency of MEN1 variants in Mexico.

In our cohort, all individuals with P/LP MEN1 variants developed panNENs and PHPT, while none of the patients with the combination of PitNET and PHPT carried MEN1 defects. As pointed out by previous studies, such cases are usually not caused by MEN1 defects, suggesting that other genetic drivers might be involved. Interestingly, 1 of these patients carried a pathogenic variant in SDHA. Germline SDHA LOF underlies the syndrome of hereditary PPGLs 5 (MIM # 614165), and less frequently causes GISTs and PitNETs, either isolated or coexisting with PPGLs (71-73). Due to low penetrance, SDHA-driven tumors are usually apparently sporadic (74). The coexistence of PHPT with acromegaly in an SDHA variant carrier has not been described before and causality for PHPT requires further confirmation. Regardless of this atypical presentation, targeted screening for additional SDHA-associated tumors will be required in this individual.

One-quarter of cases of early-onset acromegaly or gigantism, including 16.7% of FIPA, were caused by AIP LOF variants. Previous studies have reported a frequency of 29% among cases of gigantism, 6% to 20% of young sporadic PitNETs, and 15% to 20% of FIPA families (13, 38, 75-77). A single individual with MEN2A, representing one-third or the MEN2A patients in the study as well as the only 1 with familial disease, carried a well-characterized RET variant. The same defect was found in 1 patient with MTC, accounting for 12.5% of all MTC cases in the study and the only one with history of FMTC. Almost all cases of MEN2A and FMTC and around one-fifth of all cases of sporadic MTC are due to RET germline defects; variants in the same gene are found as somatic defects in around 47% of MTCs (12, 19). The 2 probands with MEN2A and negative RET screening presented sporadically. These cases could be explained by a coincidental association of NENs similar to MEN2A, or an unknown genetic syndrome.

The low frequency of SDHx variants among patients with multiple PPGLs is intriguing. Only 1 of these individuals carried an SDHD variant and had a family history of cancer predisposition. Carriers of germline LOF SDHD variants are at risk for familial predisposition to PPGLs (MIM # 168000), GISTs, renal cell carcinoma, PitNETs, and Carney–Stratakis syndrome (MIM #60686) (20, 71-73, 78, 79). The genetic origin of PPGLs involves more than 20 genes (all included in our NGS panel), but around half of cases remain without a known germline defect (23, 25). Additional unrecognized genes could be involved in the origin PPGLs in our study participants, since they belong to a population that has not been genetically characterized before. Also, no P/LP variants were identified for isolated single or multiple panNENs, multiple GI-NENs, MEN-like phenotypes, or ectopic CS. While a fraction of panNENs and GI-NENs are due germline genetic causes, ectopic CS and MEN-like are heterogeneous entities with unclear genetic backgrounds (15, 17). Additional analyses including exome/genome-wide paired sequencing of blood and tumor samples should be useful to shed light on these cases.

Our data confirm previous findings showing that NENs are part of a variety of syndromic and nonsyndromic presentations with overlapping phenotypes and genetic drivers. Nevertheless, in our cohort, three-quarters of patients with positive family history, and therefore with a high likelihood of carrying germline defects, remain without genetic diagnosis. The discovery of genetic drivers of NENs in these cases will likely require a combination of multiple technologies for genetic analyses, such as comparative genomic hybridization, and long-read exome/genome sequencing (80). Given that the number of known drivers of NENs is expected to continue growing, including the name of the affected genes in the nomenclature of these phenotypes would be advisable to improve clarity, when possible (81). This precision will become more important as strategies for personalized medicine in this field are incorporated into the clinical routine toolbox. Furthermore, future research efforts should prioritize VUS reclassification, via clinical evaluation and experimental assessment (82).

The limitations of the study are duly recognized. At this stage, our work sought to optimize the sequencing protocol and to validate the performance of the NGS panel for germline analyses. Therefore, tissue samples were analyzed for only a few cases in which either hotspot variants or LOH at the germline variant loci were investigated. As this prospective study progresses, we expect to obtain both fresh frozen tumor and blood samples from participants to perform paired analyses. This is particularly important for phenotypes that are often caused by well-characterized somatic drivers, such as MTC, PPGLs, and corticotropinomas. We are also actively recruiting additional family members, which will allow for investigating cosegregation as well as for a fine characterization of phenotype–genotype correlations. Finally, based on our results, we expect to refine the list of genes included in the panel, to maximize its diagnostic potential.

Conclusion

An NGS panel for genetic diagnosis in patients with multiple types of NENs has been designed and validated. Using this tool, we determined, for the first time, the frequency and type of genetic defects underlying multiple phenotypes of NENs in the Mexican population, including SNVs, indels, and SVs. Novel P/LP variants and VUS have been described. In addition, we described the association of a pathogenic SDHA defect with MEN1 in a single individual. Our findings will hopefully contribute to improve the access of the general population to platforms for routine genetic testing and simplify the interpretation of genetic tests in ours and other populations. Finally, our ongoing work on this prospective cohort will seek to determine how genetic variability among human populations might shape the clinical phenotypes and outcomes in patients with monogenic causes of NENs.

Acknowledgments

We are grateful for the kind collaboration of patients and their family members. Also, we would like to thank the fellows and attending clinicians who supported the project in the participating hospitals. Finally, we would like to acknowledge the kind help of Ms. Teresa Zariñán-García, MSc (Red de Apoyo a la Investigación) with Sanger sequencing and of Dr. Haydee Miranda-Ortiz (Instituto Nacional de Medicina Genómica) and Dr. Carlo César Cortés-González (Instituto Nacional de Cancerología) who provided access to ddPCR instruments.

Abbreviations

CD

Cushing's disease

CS

Cushing's syndrome

dd

droplet digital

FFPE

formalin-fixed and paraffin-embedded

FIPA

familial isolated pituitary adenoma

FMTC

familial MTC

GH

growth hormone

GI-NEN

gastrointestinal NEN

GIST

gastrointestinal stromal tumor

LB

likely benign

LOF

loss of function

LOH

loss of heterozygosity

MEN

multiple endocrine neoplasia

MTC

medullary thyroid carcinoma

NEN

neuroendocrine neoplasm

NF1

neurofibromatosis type 1

NGS

next-generation sequencing

panNEN

pancreatic NEN

PCR

polymerase chain reaction

PHPT

primary hyperparathyroidism

PitNET

pituitary neuroendocrine tumor

P/LP

pathogenic or likely pathogenic

PPGL

pheochromocytoma and paraganglioma

PTC

papillary thyroid carcinoma

SNV

single nucleotide variant

SV

structural variant

UTR

untranslated region

VHL

Von Hippel–Lindau syndrome

VUS

variant of uncertain significance

Contributor Information

Laura C Hernández-Ramírez, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Claudia Ramírez-Rentería, Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Rosa G Rebollar-Vega, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Julia M Zuarth-Vázquez, Department of Endocrinology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Mariana Torres-Morán, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Alexa L Franco-Álvarez, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Andrea I Manzo-Osnaya, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Edgar Martell-Jiménez, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Maribel Rodríguez-Torres, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Wellbert E Hernández-Núñez, Department of Endocrinology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Alfredo A Reza-Albarrán, Department of Endocrinology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Armando Gamboa-Domínguez, Departmento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Etual Espinosa-Cárdenas, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Ernesto Sosa-Eroza, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Irma Hernández-García, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Baldomero González-Virla, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Alex F Hernández-Martínez, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Manuel García-Sáenz, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Aldo Eguiluz-Melendez, Department of Neurosurgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico.

Moisés Mercado, Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Guadalupe Vargas-Ortega, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Lourdes Balcázar-Hernández, Department of Endocrinology, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.

Funding

Our research was supported by departmental funding from the Coordination of Scientific Research from Universidad Nacional Autónoma de México (UNAM), research grants from UNAM's Support Program for Research Projects and Technological Innovation (UNAM-PAPIIT, projects TA200322 and TA200524) and Sociedad Mexicana de Nutrición y Endocrinología, and an equipment grant from the United Kingdom's Society for Endocrinology.

Disclosures

The authors declare no conflicts of interest concerning the contents of this manuscript.

Data Availability

Sequencing data have been deposited to NCBI BioProject. Original deidentified research data pertaining to this study are available upon reasonable request by contacting L.C.H.R.

Ethics Declaration

Study participants provided written informed consent and were recruited under a protocol approved by the IRBs of Hospital de Especialidades de Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, respectively (protocol IDs R-2022-785-011 and 4090, respectively; ClinicalTrials.gov: NCT06523582).

Clinical Trial Information

R-2022-785-011 (Hospital de Especialidades de Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, approved March 23, 2022), 4090 (Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, approved April 13, 2022), and NCT06523582 (ClinicalTrials.gov, uploaded July 26, 2024).

References

  • 1. Johansson  E, Andersson  L, Ornros  J, et al.  Revising the embryonic origin of thyroid C cells in mice and humans. Development. 2015;142(20):3519‐3528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kulke  MH, Shah  MH, Benson  AB  3rd, et al.  National comprehensive cancer n. Neuroendocrine tumors, version 1.2015. J Natl Compr Canc Netw. 2015;13(1):78‐108. [DOI] [PubMed] [Google Scholar]
  • 3. Furlan  A, Dyachuk  V, Kastriti  ME, et al.  Multipotent peripheral glial cells generate neuroendocrine cells of the adrenal medulla. Science. 2017;357(6346):eaal3753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Kastriti  ME, Kameneva  P, Kamenev  D, et al.  Schwann cell precursors generate the majority of chromaffin cells in Zuckerkandl organ and some sympathetic neurons in paraganglia. Front Mol Neurosci. 2019;12:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Crona  J, Skogseid  B. GEP- NETS UPDATE: Genetics of neuroendocrine tumors. Eur J Endocrinol. 2016;174(6):R275‐R290. [DOI] [PubMed] [Google Scholar]
  • 6. Kloppel  G, Rindi  G, Anlauf  M, Perren  A, Komminoth  P. Site-specific biology and pathology of gastroenteropancreatic neuroendocrine tumors. Virchows Arch. 2007;451(Suppl 1):S9‐S27. [DOI] [PubMed] [Google Scholar]
  • 7. Zandee  WT, Kamp  K, van Adrichem  RC, Feelders  RA, de Herder  WW. Effect of hormone secretory syndromes on neuroendocrine tumor prognosis. Endocr Relat Cancer. 2017;24:R261‐R274. [DOI] [PubMed] [Google Scholar]
  • 8. Melmed  S. Pituitary-tumor endocrinopathies. N Engl J Med. 2020;382(10):937‐950. [DOI] [PubMed] [Google Scholar]
  • 9. Yao  JC, Hassan  M, Phan  A, et al.  One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26(18):3063‐3072. [DOI] [PubMed] [Google Scholar]
  • 10. Capdevila  J, Casanovas  O, Salazar  R, et al.  Translational research in neuroendocrine tumors: pitfalls and opportunities. Oncogene. 2017;36(14):1899‐1907. [DOI] [PubMed] [Google Scholar]
  • 11. Wu  P, He  D, Chang  H, Zhang  X. Epidemiologic trends of and factors associated with overall survival in patients with neuroendocrine tumors over the last two decades in the USA. Endocr Connect. 2023;12(12):e230331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Fagin  JA, Wells  SA  Jr. Biologic and clinical perspectives on thyroid cancer. N Engl J Med. 2016;375(23):2307. [DOI] [PubMed] [Google Scholar]
  • 13. Iacovazzo  D, Hernández-Ramírez  LC, Korbonits  M. Sporadic pituitary adenomas: the role of germline mutations and recommendations for genetic screening. Expert Rev Endocrinol Metab. 2017;12(2):143‐153. [DOI] [PubMed] [Google Scholar]
  • 14. Bilezikian  JP. Primary Hyperparathyroidism. J Clin Endocrinol Metab. 2018;103(11):3993‐4004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Mafficini  A, Scarpa  A. Genetics and epigenetics of gastroenteropancreatic neuroendocrine neoplasms. Endocr Rev. 2019;40(2):506‐536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Papathomas  TG, Suurd  DPD, Pacak  K, et al.  What have we learned from molecular biology of paragangliomas and pheochromocytomas?  Endocr Pathol. 2021;32(1):134‐153. [DOI] [PubMed] [Google Scholar]
  • 17. Perez  K, Kulke  MH, Chittenden  A, et al.  Clinical implications of pathogenic germline variants in small intestine neuroendocrine tumors (SI-NETs). JCO Precis Oncol. 2021;5(5):808‐816. [DOI] [PubMed] [Google Scholar]
  • 18. Jha  S, Simonds  WF. Molecular and clinical spectrum of primary hyperparathyroidism. Endocr Rev. 2023;44(5):779‐818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Xu  B, Viswanathan  K, Ahadi  MS, et al.  Association of the genomic profile of medullary thyroid carcinoma with tumor characteristics and clinical outcomes in an international multicenter study. Thyroid. 2024;34(2):167‐176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Pasini  B, McWhinney  SR, Bei  T, et al.  Clinical and molecular genetics of patients with the Carney-Stratakis syndrome and germline mutations of the genes coding for the succinate dehydrogenase subunits SDHB, SDHC, and SDHD. Eur J Hum Genet. 2008;16(1):79‐88. [DOI] [PubMed] [Google Scholar]
  • 21. Scarpa  A, Chang  DK, Nones  K, et al.  Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65‐71. [DOI] [PubMed] [Google Scholar]
  • 22. Pilarski  R. PTEN hamartoma tumor syndrome: a clinical overview. Cancers (Basel). 2019;11(6):844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Buffet  A, Burnichon  N, Favier  J, Gimenez-Roqueplo  AP. An overview of 20 years of genetic studies in pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab. 2020;34(2):101416. [DOI] [PubMed] [Google Scholar]
  • 24. Chevalier  B, Dupuis  H, Jannin  A, et al.  Phakomatoses and endocrine gland tumors: noteworthy and (not so) rare associations. Front Endocrinol (Lausanne). 2021;12:678869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Nölting  S, Bechmann  N, Taieb  D, et al.  Personalized management of pheochromocytoma and paraganglioma. Endocr Rev. 2022;43(2):199‐239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Singeisen  H, Renzulli  MM, Pavlicek  V, et al.  Multiple endocrine neoplasia type 4: a new member of the MEN family. Endocr Connect. 2023;12(2):e220411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Hernández-Ramírez  LC, Perez-Rivas  LG, Theodoropoulou  M, Korbonits  M. An update on the genetic drivers of corticotroph tumorigenesis. Exp Clin Endocrinol Diabetes. 2024;132(12):678‐696. [DOI] [PubMed] [Google Scholar]
  • 28. Okafor  C, Hogan  J, Raygada  M, et al.  Update on targeted therapy in medullary thyroid cancer. Front Endocrinol (Lausanne). 2021;12:708949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Torres-Morán  M, Franco-Álvarez  AL, Rebollar-Vega  RG, Hernández-Ramírez  LC. Hotspots of somatic genetic variation in pituitary neuroendocrine tumors. Cancers (Basel). 2023;15(23):5685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Lin  AL, Rudneva  VA, Richards  AL, et al.  Genome-wide loss of heterozygosity predicts aggressive, treatment-refractory behavior in pituitary neuroendocrine tumors. Acta Neuropathol. 2024;147(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Webster  AP, Thirlwell  C. The molecular biology of midgut neuroendocrine neoplasms. Endocr Rev. 2024;45(3):343‐350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Denes  J, Swords  F, Rattenberry  E, et al.  Heterogeneous genetic background of the association of pheochromocytoma/paraganglioma and pituitary adenoma: results from a large patient cohort. J Clin Endocrinol Metab. 2015;100(3):E531‐E541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Xekouki  P, Szarek  E, Bullova  P, et al.  Pituitary adenoma with paraganglioma/pheochromocytoma (3PAs) and succinate dehydrogenase defects in humans and mice. J Clin Endocrinol Metabolism. 2015;100(5):E710‐E719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Persani  L, de Filippis  T, Colombo  C, Gentilini  D. GENETICS IN ENDOCRINOLOGY: Genetic diagnosis of endocrine diseases by NGS: novel scenarios and unpredictable results and risks. Eur J Endocrinol. 2018;179(3):R111‐R123. [DOI] [PubMed] [Google Scholar]
  • 35. Seabrook  AJ, Harris  JE, Velosa  SB, et al.  Multiple endocrine tumors associated with germline MAX mutations: multiple endocrine neoplasia type 5?  J Clin Endocrinol Metab. 2021;106(4):1163‐1182. [DOI] [PubMed] [Google Scholar]
  • 36. Seabrook  A, Wijewardene  A, De Sousa  S, et al.  MEN4, the MEN1 mimicker: a case series of three phenotypically heterogenous patients with unique CDKN1B mutations. J Clin Endocrinol Metab. 2022;107(8):2339‐2349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Thakker  RV, Newey  PJ, Walls  GV, et al.  Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012;97(9):2990‐3011. [DOI] [PubMed] [Google Scholar]
  • 38. Hernández-Ramírez  LC, Gabrovska  P, Dénes  J, et al.  Landscape of familial isolated and young-onset pituitary adenomas: prospective diagnosis in AIP mutation carriers. J Clin Endocrinol Metab. 2015;100(9):E1242‐E1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wells  SA  Jr, Asa  SL, Dralle  H, et al.  American thyroid association guidelines task force on medullary thyroid C. Revised American thyroid association guidelines for the management of medullary thyroid carcinoma. Thyroid. 2015;25(6):567‐610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Silva-Zolezzi  I, Hidalgo-Miranda  A, Estrada-Gil  J, et al.  Analysis of genomic diversity in Mexican mestizo populations to develop genomic medicine in Mexico. Proc Natl Acad Sci U S A  2009; 106(21):8611‐8616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Ziyatdinov  A, Torres  J, Alegre-Diaz  J, et al.  Genotyping, sequencing and analysis of 140,000 adults from Mexico City. Nature. 2023;622(7984):784‐793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Hernández-Ramírez  LC, Ramírez-Rentería  C, Rebollar-Vega  RG, et al.  Supplementary material to: overlapping presentations and diverse genetic defects characterize neuroendocrine neoplasms in a Mexican cohort. 2025. Accessed January 16, 2025. 10.5281/zenodo.14676467 [DOI] [PMC free article] [PubMed]
  • 43. Babraham Bioinformatics. FastQC: a quality control tool for high throughput sequence data. 2010. Accessed September 20, 2024. http://www.bioinformatics.babraham.ac.uk/projects/fastqc/
  • 44. Li  H, Durbin  R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26(5):589‐595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Broad Institute. Picard. Broad Institute, GitHub repository. Accessed September 20, 2024. https://broadinstitute.github.io/picard/
  • 46. Broad Institute. FreeBayes. Broad Institute, GitHub repository. Accessed September 20, 2024. https://github.com/freebayes/freebayes
  • 47. Plagnol  V, Curtis  J, Epstein  M, et al.  A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics. 2012;28(21):2747‐2754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Genoox. Franklin. Accessed September 20, 2024. https://franklin.genoox.com/clinical-db/home
  • 49. Chen  S, Francioli  LC, Goodrich  JK, et al.  A genomic mutational constraint map using variation in 76,156 human genomes. Nature. 2024;625(7993):92‐100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. The All of Us Research Program Genomics Investigators . Genomic data in the all of Us research program. Nature. 2024;627(8003):340‐346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Robinson  JT, Thorvaldsdottir  H, Winckler  W, et al.  Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Landrum  MJ, Lee  JM, Riley  GR, et al.  ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 2014;42(D1):D980‐D985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Richards  S, Aziz  N, Bale  S, et al.  Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology. Genet Med. 2015;17(5):405‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Kopanos  C, Tsiolkas  V, Kouris  A, et al.  VarSome: the human genomic variant search engine. Bioinformatics. 2019;35(11):1978‐1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Chunn  LM, Nefcy  DC, Scouten  RW, et al.  Mastermind: a comprehensive genomic association search engine for empirical evidence curation and genetic variant interpretation. Front Genet. 2020;11:577152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Hart  RK, Fokkema  I, DiStefano  M, et al.  HGVS nomenclature 2024: improvements to community engagement, usability, and computability. Genome Med. 2024;16(1):149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Bouras  A, Lefol  C, Ruano  E, Grand-Masson  C, Wang  Q. PMS2 or PMS2CL? Characterization of variants detected in the 3′ of the PMS2 gene. Genes Chromosomes Cancer. 2024;63(1):e23193. [DOI] [PubMed] [Google Scholar]
  • 58. Elbehti-Green  A, Au  HC, Mascarello  JT, Ream-Robinson  D, Scheffler  IE. Characterization of the human SDHC gene encoding of the integral membrane proteins of succinate-quinone oxidoreductase in mitochondria. Gene. 1998;213(1-2):133‐140. [DOI] [PubMed] [Google Scholar]
  • 59. De Sousa  SM, McCabe  MJ, Wu  K, et al.  Germline variants in familial pituitary tumour syndrome genes are common in young patients and families with additional endocrine tumours. Eur J Endocrinol. 2017;176(5):635‐644. [DOI] [PubMed] [Google Scholar]
  • 60. Ramírez-Rentería  C, Hernández-Ramírez  LC, Portocarrero-Ortiz  L, et al.  AIP mutations in young patients with acromegaly and the Tampico Giant: the Mexican experience. Endocrine. 2016;53(2):402‐411. [DOI] [PubMed] [Google Scholar]
  • 61. Hernández-Ramírez  LC, Gam  R, Valdés  N, et al.  Loss-of-function mutations in the CABLES1 gene are a novel cause of Cushing's disease. Endocr Relat Cancer. 2017;24(8):379‐392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Andreou  A, Yngvadottir  B, Bassaganyas  L, et al.  Elongin C (ELOC/TCEB1)-associated von Hippel-Lindau disease. Hum Mol Genet. 2022;31(16):2728‐2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Teh  BT, Zedenius  J, Kytola  S, et al.  Thymic carcinoids in multiple endocrine neoplasia type 1. Ann Surg. 1998;228(1):99‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Nachtigall  LB, Guarda  FJ, Lines  KE, et al.  Clinical MEN-1 among a large cohort of patients with acromegaly. J Clin Endocrinol Metab. 2020;105(6):e2271‐e2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Lavezzi  E, Brunetti  A, Smiroldo  V, et al.  Case report: new CDKN1B mutation in multiple endocrine neoplasia type 4 and brief literature review on clinical management. Front Endocrinol (Lausanne). 2022;13:773143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Bean  LJH, Funke  B, Carlston  CM, et al.  Diagnostic gene sequencing panels: from design to report-a technical standard of the American college of medical genetics and genomics (ACMG). Genet Med. 2020;22(3):453‐461. [DOI] [PubMed] [Google Scholar]
  • 67. Cebrian  A, Ruiz-Llorente  S, Cascon  A, et al.  Mutational and gross deletion study of the MEN1 gene and correlation with clinical features in Spanish patients. J Med Genet. 2003;40(5):e72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Lemos  MC, Thakker  RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008;29(1):22‐32. [DOI] [PubMed] [Google Scholar]
  • 69. de Laat  JM, van der Luijt  RB, Pieterman  CR, et al.  MEN1 redefined, a clinical comparison of mutation-positive and mutation-negative patients. BMC Med. 2016;14(1):182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Romanet  P, Mohamed  A, Giraud  S, et al.  UMD-MEN1 database: an overview of the 370 MEN1 variants present in 1676 patients from the French population. J Clin Endocrinol Metab. 2019;104(3):753‐764. [DOI] [PubMed] [Google Scholar]
  • 71. Burnichon  N, Briere  JJ, Libe  R, et al.  SDHA is a tumor suppressor gene causing paraganglioma. Hum Mol Genet. 2010;19(15):3011‐3020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Papathomas  TG, Gaal  J, Corssmit  EP, et al.  Non-pheochromocytoma (PCC)/paraganglioma (PGL) tumors in patients with succinate dehydrogenase-related PCC-PGL syndromes: a clinicopathological and molecular analysis. Eur J Endocrinol. 2014;170(1):1‐12. [DOI] [PubMed] [Google Scholar]
  • 73. Mannelli  M, Canu  L, Ercolino  T, et al.  DIAGNOSIS of ENDOCRINE DISEASE: SDHx mutations: beyond pheochromocytomas and paragangliomas. Eur J Endocrinol. 2018;178(1):R11‐R17. [DOI] [PubMed] [Google Scholar]
  • 74. Hanson  H, Durkie  M, Lalloo  F, et al.  UK recommendations for SDHA germline genetic testing and surveillance in clinical practice. J Med Genet. 2023;60(2):107‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Daly  AF, Vanbellinghen  JF, Khoo  SK, et al.  Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92(5):1891‐1896. [DOI] [PubMed] [Google Scholar]
  • 76. Cazabat  L, Bouligand  J, Salenave  S, et al.  Germline AIP mutations in apparently sporadic pituitary adenomas: prevalence in a prospective single-center cohort of 443 patients. J Clin Endocrinol Metab. 2012;97(4):E663‐E670. [DOI] [PubMed] [Google Scholar]
  • 77. Rostomyan  L, Daly  AF, Petrossians  P, et al.  Clinical and genetic characterization of pituitary gigantism: an international collaborative study in 208 patients. Endocr Relat Cancer. 2015;22(5):745‐757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Baysal  BE, Ferrell  RE, Willett-Brozick  JE, et al.  Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science. 2000;287(5454):848‐851. [DOI] [PubMed] [Google Scholar]
  • 79. McWhinney  SR, Pasini  B, Stratakis  CA. Familial gastrointestinal stromal tumors and germ-line mutations. N Engl J Med. 2007;357(10):1054‐1056. [DOI] [PubMed] [Google Scholar]
  • 80. Kernohan  KD, Boycott  KM. The expanding diagnostic toolbox for rare genetic diseases. Nat Rev Genet. 2024;25(6):401‐415. [DOI] [PubMed] [Google Scholar]
  • 81. Biesecker  LG, Adam  MP, Alkuraya  FS, et al.  A dyadic approach to the delineation of diagnostic entities in clinical genomics. Am J Hum Genet. 2021;108(1):8‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Fowler  DM, Rehm  HL. Will variants of uncertain significance still exist in 2030?  Am J Hum Genet. 2024;111(1):5‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Hernández-Ramírez  LC. Chapter 6—the role of the aryl hydrocarbon receptor interacting protein in pituitary tumorigenesis. In: Stratakis  CA, ed. Gigantism and Acromegaly. ed. Academic Press; 2021:89‐126. [Google Scholar]
  • 84. Tanaka  C, Yoshimoto  K, Yamada  S, et al.  Absence of germ-line mutations of the multiple endocrine neoplasia type 1 (MEN1) gene in familial pituitary adenoma in contrast to MEN1 in Japanese. J Clin Endocrinol Metab. 1998;83(3):960‐965. [DOI] [PubMed] [Google Scholar]
  • 85. Vatsyayan  A, Imran  M, Bhardwaj  J, et al.  Understanding the variant landscape, and genetic epidemiology of multiple endocrine neoplasia in India. Endocrine. 2024;86(3):1178‐1187. [DOI] [PubMed] [Google Scholar]
  • 86. Kervarrec  T, Westphal  D, Pissaloux  D, et al.  Porocarcinomas with PAK1/2/3 fusions: a series of 12 cases. Histopathology. 2024;85(4):566‐578. [DOI] [PubMed] [Google Scholar]
  • 87. De Sousa  SMC, McCormack  A, Orsmond  A, et al.  Increased prevalence of germline pathogenic CHEK2 variants in individuals with pituitary adenomas. J Clin Endocrinol Metab. 2024;109(11):2720‐2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Teh  BT, Kytola  S, Farnebo  F, et al.  Mutation analysis of the MEN1 gene in multiple endocrine neoplasia type 1, familial acromegaly and familial isolated hyperparathyroidism. J Clin Endocrinol Metab. 1998;83(8):2621‐2626. [DOI] [PubMed] [Google Scholar]
  • 89. Wautot  V, Vercherat  C, Lespinasse  J, et al.  Germline mutation profile of MEN1 in multiple endocrine neoplasia type 1: search for correlation between phenotype and the functional domains of the MEN1 protein. Hum Mutat. 2002;20(1):35‐47. [DOI] [PubMed] [Google Scholar]
  • 90. Cardinal  JW, Bergman  L, Hayward  N, et al.  A report of a national mutation testing service for the MEN1 gene: clinical presentations and implications for mutation testing. J Med Genet. 2005;42(1):69‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Nord  B, Platz  A, Smoczynski  K, et al.  Malignant melanoma in patients with multiple endocrine neoplasia type 1 and involvement of the MEN1 gene in sporadic melanoma. Int J Cancer. 2000;87(4):463‐467. [DOI] [PubMed] [Google Scholar]
  • 92. Mohamed  A, Romano  D, Saveanu  A, et al.  Anti-proliferative and anti-secretory effects of everolimus on human pancreatic neuroendocrine tumors primary cultures: is there any benefit from combination with somatostatin analogs?  Oncotarget. 2017;8(25):41044‐41063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Damjanovic  SS, Antic  JA, Elezovic-Kovacevic  VI, et al.  ARMC5 alterations in patients with sporadic neuroendocrine tumors and multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2020;105(12):e4531‐e4542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Concolino  P, Costella  A, Capoluongo  E. Multiple endocrine neoplasia type 1 (MEN1): an update of 208 new germline variants reported in the last nine years. Cancer Genet. 2016;209(1-2):36‐41. [DOI] [PubMed] [Google Scholar]
  • 95. Fahsold  R, Hoffmeyer  S, Mischung  C, et al.  Minor lesion mutational spectrum of the entire NF1 gene does not explain its high mutability but points to a functional domain upstream of the GAP-related domain. Am J Hum Genet. 2000;66(3):790‐818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Valero  MC, Martin  Y, Hernandez-Imaz  E, et al.  A highly sensitive genetic protocol to detect NF1 mutations. J Mol Diagn. 2011;13(2):113‐122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Lyskjaer  I, Lindsay  D, Tirabosco  R, et al.  H3k27me3 expression and methylation status in histological variants of malignant peripheral nerve sheath tumours. J Pathol. 2020;252(2):151‐164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. de Witte  CJ, Kutzera  J, van Hoeck  A, et al.  Distinct genomic profiles are associated with treatment response and survival in ovarian cancer. Cancers (Basel). 2022;14(6):1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Sakamoto  I, Hirotsu  Y, Amemiya  K, Nozaki  T, Mochizuki  H, Omata  M. Elucidation of genomic origin of synchronous endometrial and ovarian cancer (SEO) by genomic and microsatellite analysis. J Gynecol Oncol. 2023;34(1):e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Ito  S, Iwashita  T, Asai  N, et al.  Biological properties of Ret with cysteine mutations correlate with multiple endocrine neoplasia type 2A, familial medullary thyroid carcinoma, and Hirschsprung's disease phenotype. Cancer Res. 1997;57:2870‐2872. [PubMed] [Google Scholar]
  • 101. Chappuis-Flament  S, Pasini  A, De Vita  G, et al.  Dual effect on the RET receptor of MEN 2 mutations affecting specific extracytoplasmic cysteines. Oncogene. 1998;17(22):2851‐2861. [DOI] [PubMed] [Google Scholar]
  • 102. Eng  C, Plitt  G.  Multiple endocrine neoplasia type 2. In: Adam  MP, Mirzaa  GM, Pagon  RA, eds. GeneReviews [Internet]. (WA: ): University of Washington, Seattle; 1999.  [updated 2023]. [PubMed] [Google Scholar]
  • 103. Romei  C, Mariotti  S, Fugazzola  L, et al.  Multiple endocrine neoplasia type 2 syndromes (MEN 2): results from the ItaMEN network analysis on the prevalence of different genotypes and phenotypes. Eur J Endocrinol. 2010;163(2):301‐308. [DOI] [PubMed] [Google Scholar]
  • 104. Frank-Raue  K, Rybicki  LA, Erlic  Z, et al.  Risk profiles and penetrance estimations in multiple endocrine neoplasia type 2A caused by germline RET mutations located in exon 10. Hum Mutat. 2011;32(1):51‐58. [DOI] [PubMed] [Google Scholar]
  • 105. Neocleous  V, Skordis  N, Portides  G, et al.  RET proto-oncogene mutations are restricted to codon 618 in Cypriot families with multiple endocrine neoplasia 2. J Endocrinol Invest. 2011;34(10):764‐769. [DOI] [PubMed] [Google Scholar]
  • 106. Peretz  H, Luboshitsky  R, Baron  E, et al.  Cys 618 Arg mutation in the RET proto-oncogene associated with familial medullary thyroid carcinoma and maternally transmitted Hirschsprung's disease suggesting a role for imprinting. Hum Mutat. 1997;10(2):155‐159. [DOI] [PubMed] [Google Scholar]
  • 107. Bausch  B, Schiavi  F, Ni  Y, et al.  Clinical characterization of the pheochromocytoma and paraganglioma susceptibility genes SDHA, TMEM127, MAX, and SDHAF2 for gene-informed prevention. JAMA Oncol. 2017;3(9):1204‐1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Kaplan  AI, Dwight  T, Luxford  C, Benn  DE, Clifton-Bligh  RJ. SDHA related phaeochromocytoma and paraganglioma: review and clinical management. Endocr Relat Cancer. 2024;31(10):e240111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Sen  I, Young  WF, Jr., Kasperbauer  JL, et al.  Tumor-specific prognosis of mutation-positive patients with head and neck paragangliomas. J Vasc Surg. 2020;71(5):1602‐1612.e2. [DOI] [PubMed] [Google Scholar]
  • 110. Otani  N, Sugano  K, Inami  S, et al.  Cardiac paraganglioma with a novel germline mutation of succinate dehydrogenase gene D. Jpn J Clin Oncol. 2017;47(12):1193‐1197. [DOI] [PubMed] [Google Scholar]
  • 111. Glavac  D, Neumann  HP, Wittke  C, et al.  Mutations in the VHL tumor suppressor gene and associated lesions in families with von Hippel-Lindau disease from central Europe. Hum Genet. 1996;98(3):271‐280. [DOI] [PubMed] [Google Scholar]
  • 112. Zbar  B, Kishida  T, Chen  F, et al.  Germline mutations in the Von Hippel-Lindau disease (VHL) gene in families from North America, Europe, and Japan. Hum Mutat. 1996;8(4):348‐357. [DOI] [PubMed] [Google Scholar]
  • 113. Glasker  S, Bender  BU, Apel  TW, et al.  The impact of molecular genetic analysis of the VHL gene in patients with haemangioblastomas of the central nervous system. J Neurol Neurosurg Psychiatry. 1999;67(6):758‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Webster  AR, Maher  ER, Moore  AT. Clinical characteristics of ocular angiomatosis in von Hippel-Lindau disease and correlation with germline mutation. Arch Ophthalmol. 1999;117(3):371‐378. [DOI] [PubMed] [Google Scholar]
  • 115. Ong  KR, Woodward  ER, Killick  P, Lim  C, Macdonald  F, Maher  ER. Genotype-phenotype correlations in von Hippel-Lindau disease. Hum Mutat. 2007;28(2):143‐149. [DOI] [PubMed] [Google Scholar]
  • 116. Perigny  M, Hammel  P, Corcos  O, et al.  Pancreatic endocrine microadenomatosis in patients with von Hippel-Lindau disease: characterization by VHL/HIF pathway proteins expression. Am J Surg Pathol. 2009;33(5):739‐748. [DOI] [PubMed] [Google Scholar]
  • 117. Erlic  Z, Ploeckinger  U, Cascon  A, et al.  Systematic comparison of sporadic and syndromic pancreatic islet cell tumors. Endocr Relat Cancer. 2010;17(4):875‐883. [DOI] [PubMed] [Google Scholar]
  • 118. Rasmussen  A, Alonso  E, Ochoa  A, et al.  Uptake of genetic testing and long-term tumor surveillance in von Hippel-Lindau disease. BMC Med Genet. 2010;11(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Bausch  B, Wellner  U, Peyre  M, et al.  International endolymphatic sac tumor C. Characterization of endolymphatic sac tumors and von Hippel-Lindau disease in the international endolymphatic sac tumor registry. Head Neck. 2016;38(S1):E673‐E679. [DOI] [PubMed] [Google Scholar]
  • 120. Krauss  T, Ferrara  AM, Links  TP, et al.  Preventive medicine of von Hippel-Lindau disease-associated pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2018;25(9):783‐793. [DOI] [PubMed] [Google Scholar]
  • 121. Hong  B, Ma  K, Zhou  J, et al.  Frequent mutations of VHL gene and the clinical phenotypes in the largest Chinese cohort with Von Hippel-Lindau disease. Front Genet. 2019;10:867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Hajjaj  A, van Overdam  KA, Oldenburg  RA, et al.  Retinal haemangioblastomas in von Hippel-Lindau germline mutation carriers: progression, complications and treatment outcome. Acta Ophthalmol. 2020;98(5):464‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Reich  M, Jaegle  S, Neumann-Haefelin  E, et al.  Genotype-phenotype correlation in von Hippel-Lindau disease. Acta Ophthalmol. 2021;99(8):e1492‐e1500. [DOI] [PubMed] [Google Scholar]
  • 124. Chacon-Camacho  OF, Rodriguez-Dennen  F, Camacho-Molina  A, Rasmussen  A, Alonso-Vilatela  E, Zenteno  JC. Clinical and molecular features of familial and sporadic cases of von Hippel-Lindau disease from Mexico. Clin Exp Ophthalmol. 2010;38(3):277‐283. [DOI] [PubMed] [Google Scholar]
  • 125. Lei  Z, Klasson  TD, Brandt  MM, et al.  Control of angiogenesis via a VHL/miR-212/132 axis. Cells. 2020;9(4):1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Cherkasova  E, Malinzak  E, Rao  S, et al.  Inactivation of the von Hippel-Lindau tumor suppressor leads to selective expression of a human endogenous retrovirus in kidney cancer. Oncogene. 2011;30(47):4697‐4706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. von Teichman  A, Storz  M, Dettwiler  S, Moch  H, Schraml  P. Whole genome and transcriptome amplification: practicable tools for sustainable tissue biobanking?  Virchows Arch. 2012;461(5):571‐580. [DOI] [PubMed] [Google Scholar]
  • 128. Hahn  AW, Gill  DM, Maughan  B, et al.  Correlation of genomic alterations assessed by next-generation sequencing (NGS) of tumor tissue DNA and circulating tumor DNA (ctDNA) in metastatic renal cell carcinoma (mRCC): potential clinical implications. Oncotarget. 2017;8(20):33614‐33620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Pilling  A, Wee  C, Bar-Meir  E, et al.  The potential and limitations of precision oncology: lessons learned from whole-exome sequencing in an exceptional response to Everolimus in advanced renal cell carcinoma. Case Rep Oncol. 2021;14(2):1194‐1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Cybulski  C, Matyjasik  J, Soroka  M, et al.  Mutations in the von hippel-lindau tumour suppressor gene in central nervous system hemangioblastomas. Hered Cancer Clin Pract. 2004;2(2):93‐97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Rebollar-Vega  RG, Zuarth-Vázquez  JM, Hernández-Ramírez  LC. Clinical spectrum of USP8 pathogenic variants in Cushing's disease. Arch Med Res. 2023;54(8):102899. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Citations

  1. Hernández-Ramírez  LC, Ramírez-Rentería  C, Rebollar-Vega  RG, et al.  Supplementary material to: overlapping presentations and diverse genetic defects characterize neuroendocrine neoplasms in a Mexican cohort. 2025. Accessed January 16, 2025. 10.5281/zenodo.14676467 [DOI] [PMC free article] [PubMed]

Data Availability Statement

Sequencing data have been deposited to NCBI BioProject. Original deidentified research data pertaining to this study are available upon reasonable request by contacting L.C.H.R.


Articles from The Journal of Clinical Endocrinology and Metabolism are provided here courtesy of The Endocrine Society

RESOURCES