1
|
Hernández-Ramírez LC, Perez-Rivas LG, Theodoropoulou M, Korbonits M. An Update on the Genetic Drivers of Corticotroph Tumorigenesis. Exp Clin Endocrinol Diabetes 2024; 132:678-696. [PMID: 38830604 DOI: 10.1055/a-2337-2265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
The genetic landscape of corticotroph tumours of the pituitary gland has dramatically changed over the last 10 years. Somatic changes in the USP8 gene account for the most common genetic defect in corticotrophinomas, especially in females, while variants in TP53 or ATRX are associated with a subset of aggressive tumours. Germline defects have also been identified in patients with Cushing's disease: some are well-established (MEN1, CDKN1B, DICER1), while others are rare and could represent coincidences. In this review, we summarise the current knowledge on the genetic drivers of corticotroph tumorigenesis, their molecular consequences, and their impact on the clinical presentation and prognosis.
Collapse
Affiliation(s)
- 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, Mexico
| | | | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, Munich 80336, Germany
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
2
|
Petzold LM, Busse M, Mohr H, Pellegata NS, Pfeiffer F, Herzen J. Development of a H&E multi-agent-staining method for laboratory-microCT applied to endocrine glands of MENX rats. DEVELOPMENTS IN X-RAY TOMOGRAPHY XV 2024:3. [DOI: 10.1117/12.3029584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Lanzaro F, De Biasio D, Cesaro FG, Stampone E, Tartaglione I, Casale M, Bencivenga D, Marzuillo P, Roberti D. Childhood Multiple Endocrine Neoplasia (MEN) Syndromes: Genetics, Clinical Heterogeneity and Modifying Genes. J Clin Med 2024; 13:5510. [PMID: 39336996 PMCID: PMC11432259 DOI: 10.3390/jcm13185510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/15/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple endocrine neoplasia (MEN) syndromes are part of a spectrum of clinically well-defined tumor syndromes ultimately characterized by histologically similar tumors arising in patients and families with mutations in one of the following four genes: MEN1, RET, CDKN1B, and MAX. The high level of genetic and phenotypic heterogeneity has been linked to phenocopies and modifying genes, as well as unknown mechanisms that might be investigated in the future based on preclinical and translational considerations. MEN1, also known as Wermer's syndrome (OMIM *131100), is an autosomal dominant syndrome codifying for the most frequent MEN syndrome showing high penetrance due to mutations in the MEN1 gene; nevertheless, clinical manifestations vary among patients in terms of tumor localization, age of onset, and clinical aggressiveness/severity, even within the same families. This has been linked to the effect of modifying genes, as described in the review. MEN 2-2b-4 and 5 also show remarkable clinical heterogeneity. The traditional view of genetically predisposing monogenic or multifactorial disorders is no longer valid, and mandates a change in scientific focus. Phenotypes are indeed rarely consistent across genetic backgrounds and environments. In the future, understanding factors and genetic variants that control cellular functions and the expression of disease genes should provide insights into fundamental disease processes, providing implications for counseling and therapeutic and prophylactic possibilities.
Collapse
Affiliation(s)
- Francesca Lanzaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Delia De Biasio
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Francesco Giustino Cesaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Emanuela Stampone
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Immacolata Tartaglione
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Maddalena Casale
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Domenico Roberti
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| |
Collapse
|
4
|
Cetani F, Dinoi E, Pierotti L, Pardi E. Familial states of primary hyperparathyroidism: an update. J Endocrinol Invest 2024; 47:2157-2176. [PMID: 38635114 DOI: 10.1007/s40618-024-02366-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/24/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Familial primary hyperparathyroidism (PHPT) includes syndromic and non-syndromic disorders. The former are characterized by the occurrence of PHPT in association with extra-parathyroid manifestations and includes multiple endocrine neoplasia (MEN) types 1, 2, and 4 syndromes, and hyperparathyroidism-jaw tumor (HPT-JT). The latter consists of familial hypocalciuric hypercalcemia (FHH) types 1, 2 and 3, neonatal severe primary hyperparathyroidism (NSHPT), and familial isolated primary hyperparathyroidism (FIHP). The familial forms of PHPT show different levels of PHPT penetrance, developing earlier and with multiglandular involvement compared to sporadic counterpart. All these diseases exhibit Mendelian inheritance patterns, and for most of them, the genes responsible have been identified. DNA testing for predisposing mutations is helpful in index cases or in individuals with a high suspicion of the disease. Early recognition of hereditary disorders of PHPT is of great importance for the best clinical and surgical approach. Genetic testing is useful in routine clinical practice because it will also involve appropriate screening for extra-parathyroidal manifestations related to the syndrome as well as the identification of asymptomatic carriers of the mutation. PURPOSE The aim of the review is to discuss the current knowledge on the clinical and genetic profile of these disorders along with the importance of genetic testing in clinical practice.
Collapse
Affiliation(s)
- F Cetani
- Endocrine Unit 2, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - E Dinoi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L Pierotti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - E Pardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Balinisteanu I, Caba L, Florea A, Popescu R, Florea L, Ungureanu MC, Leustean L, Gorduza EV, Preda C. Unlocking the Genetic Secrets of Acromegaly: Exploring the Role of Genetics in a Rare Disorder. Curr Issues Mol Biol 2024; 46:9093-9121. [PMID: 39194755 DOI: 10.3390/cimb46080538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/29/2024] Open
Abstract
Acromegaly is a rare endocrine disorder characterized by the excessive production of growth hormone (GH) in adulthood. Currently, it is understood that certain pituitary neuroendocrine tumors (PitNETs) exhibit a hereditary predisposition. These tumors' genetic patterns fall into two categories: isolated and syndromic tumors. The isolated forms are characterized by molecular defects that predispose exclusively to PitNETs, including familial isolated pituitary adenomas (FIPAs) and sporadic genetic defects not characterized by hereditary predisposition. All the categories involve either germline or somatic mutations, or both, each associated with varying levels of penetrance and different phenotypes. This highlights the importance of genetic testing and the need for a more comprehensive view of the whole disease. Despite the availability of multiple treatment options, diagnosis often occurs after several years, and management is still difficult. Early detection and intervention are crucial for preventing complications and enhancing the quality of life for affected individuals. This review aims to elucidate the molecular, clinical, and histological characteristics of GH-secreting PitNETs, providing insights into their prevalence, treatment nuances, and the benefits of genetic testing for each type of genetic disorder associated with acromegaly.
Collapse
Affiliation(s)
- Ioana Balinisteanu
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Lavinia Caba
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Andreea Florea
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Roxana Popescu
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Florea
- Nephrology-Internal Medicine Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Maria-Christina Ungureanu
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Letitia Leustean
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Preda
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
6
|
Martinerie L, Bouligand J, North MO, Bertherat J, Assié G, Espiard S. Consensus statement by the French Society of Endocrinology (SFE) and French Society of Pediatric Endocrinology & Diabetology (SFEDP) for the diagnosis of Cushing's syndrome: Genetics of Cushing's syndrome. ANNALES D'ENDOCRINOLOGIE 2024; 85:284-293. [PMID: 38253221 DOI: 10.1016/j.ando.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
Cushing's syndrome is due to overproduction of cortisol, leading to abnormal and prolonged exposure to cortisol. The most common etiology is Cushing disease, while adrenal causes are rarer. Knowledge of the genetics of Cushing's syndrome, and particularly the adrenal causes, has improved considerably over the last 10 years, thanks in particular to technical advances in high-throughput sequencing. The present study, by a group of experts from the French Society of Endocrinology and the French Society of Pediatric Endocrinology and Diabetology, reviewed the literature on germline genetic alterations leading to a predisposition to develop Cushing's syndrome. The review led to a consensus statement on genetic screening for Cushing disease and adrenal Cushing's syndrome.
Collapse
Affiliation(s)
- Laetitia Martinerie
- Department of Pediatric Endocrinology, CHU Robert-Debré, AP-HP, Paris, France
| | - Jérôme Bouligand
- Faculté de médecine Paris-Saclay, Inserm Unit UMRS1185 Endocrine Physiology and Physiopathology, Paris, France
| | - Marie-Odile North
- Department of Genetics and Molecular Biology, hôpital Cochin, AP-HP, University of Paris, Paris, France
| | - Jérôme Bertherat
- Endocrinology Department, centre de référence maladies rares de la surrénale (CRMRS), hôpital Cochin, AP-HP, University of Paris, Paris, France
| | - Guillaume Assié
- Endocrinology Department, centre de référence maladies rares de la surrénale (CRMRS), hôpital Cochin, AP-HP, University of Paris, Paris, France
| | - Stéphanie Espiard
- Service d'endocrinologie, diabétologie, métabolisme et nutrition, CHU de Lille, 59000 Lille, France.
| |
Collapse
|
7
|
Chevalier B, Coppin L, Romanet P, Cuny T, Maïza JC, Abeillon J, Forestier J, Walter T, Gilly O, Le Bras M, Smati S, Nunes ML, Geslot A, Grunenwald S, Mouly C, Arnault G, Wagner K, Koumakis E, Cortet-Rudelli C, Merlen É, Jannin A, Espiard S, Morange I, Baudin É, Cavaille M, Tauveron I, Teissier MP, Borson-Chazot F, Mirebeau-Prunier D, Savagner F, Pasmant É, Giraud S, Vantyghem MC, Goudet P, Barlier A, Cardot-Bauters C, Odou MF. Beyond MEN1, When to Think About MEN4? Retrospective Study on 5600 Patients in the French Population and Literature Review. J Clin Endocrinol Metab 2024; 109:e1482-e1493. [PMID: 38288531 DOI: 10.1210/clinem/dgae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/18/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024]
Abstract
CONTEXT Germline CDKN1B variants predispose patients to multiple endocrine neoplasia type 4 (MEN4), a rare MEN1-like syndrome, with <100 reported cases since its discovery in 2006. Although CDKN1B mutations are frequently suggested to explain cases of genetically negative MEN1, the prevalence and phenotype of MEN4 patients is poorly known, and genetic counseling is unclear. OBJECTIVE To evaluate the prevalence of MEN4 in MEN1-suspected patients and characterize the phenotype of MEN4 patients. DESIGN Retrospective observational nationwide study. Narrative review of literature and variant class reassessment. PATIENTS We included all adult patients with class 3/4/5 CDKN1B variants identified by the laboratories from the French Oncogenetic Network on Neuroendocrine Tumors network between 2015 and 2022 through germline genetic testing for MEN1 suspicion. After class reassessment, we compared the phenotype of symptomatic patients with class 4/5 CDKN1B variants (ie, with genetically confirmed MEN4 diagnosis) in our series and in literature with 66 matched MEN1 patients from the UMD-MEN1 database. RESULTS From 5600 MEN1-suspected patients analyzed, 4 with class 4/5 CDKN1B variant were found (0.07%). They presented with multiple duodenal NET, primary hyperparathyroidism (PHPT) and adrenal nodule, isolated PHPT, PHPT, and pancreatic neuroendocrine tumor. We listed 29 patients with CDKN1B class 4/5 variants from the literature. Compared with matched MEN1 patients, MEN4 patients presented lower NET incidence and older age at PHPT diagnosis. CONCLUSION The prevalence of MEN4 is low. PHPT and pituitary adenoma represent the main associated lesions, NETs are rare. Our results suggest a milder and later phenotype than in MEN1. Our observations will help to improve genetic counseling and management of MEN4 families.
Collapse
Affiliation(s)
- Benjamin Chevalier
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
- University of Lille, 59000 Lille, France
- Department of Nuclear Medicine, Lille University Hospital, 59000 Lille, France
| | - Lucie Coppin
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer-Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
- CHU Lille, Service de Biochimie et Biologie moléculaire « Hormonologie, Métabolisme-Nutrition, Oncologie, 59000 Lille, France
| | - Pauline Romanet
- Laboratory of Molecular Biology GEnOPé, Biogénopôle, Aix Marseille Univ, APHM, INSERM, UMR1251 MMG, Hôpital de la Timone, 13005 Marseille, France
| | - Thomas Cuny
- Department of Endocrinology, Aix Marseille Univ, APHM, INSERM, UMR1251 MMG, MARMARA Institute, CRMR HYPO, Hôpital de la Conception, 13005 Marseille, France
| | - Jean-Christophe Maïza
- Department of Endocrinology, Diabetes, and Nutrition, GHSR, Centre Hospitalo-Universitaire de la Réunion, 97448 Saint-Pierre, La Réunion, France
| | - Juliette Abeillon
- Hospices Civils de Lyon, Fédération d'Endocrinologie, Université Claude Bernard Lyon 1, 69003 Lyon, France
| | - Julien Forestier
- Service d'Oncologie Médicale et Hépatogastroentérologie, Hospices Civil de Lyon, 69003 Lyon, France
| | - Thomas Walter
- Service d'Oncologie Médicale et Hépatogastroentérologie, Hospices Civil de Lyon, 69003 Lyon, France
- Université de Lyon, 69003 Lyon, France
| | - Olivier Gilly
- Department of Metabolic and Endocrine Disease, CHU Nîmes, Université Montpellier, 30900 Nîmes, France
| | - Maëlle Le Bras
- Service d'endocrinologie, diabétologie, nutrition, Nantes Université, CHU Nantes, l'institut du thorax, F-44000 Nantes, France
| | - Sarra Smati
- Service d'endocrinologie, diabétologie, nutrition, Nantes Université, CHU Nantes, l'institut du thorax, F-44000 Nantes, France
| | - Marie Laure Nunes
- Department of Endocrinology, Diabetes and Nutrition, University Hospital (CHU) and University of Bordeaux, 33404 Bordeaux, France
| | - Aurore Geslot
- Service d'endocrinologie, maladies métaboliques et nutrition, pôle cardio-vasculaire et métabolique, CHU Larrey, 31059 Toulouse cedex, France
| | - Solange Grunenwald
- Service d'endocrinologie, maladies métaboliques et nutrition, pôle cardio-vasculaire et métabolique, CHU Larrey, 31059 Toulouse cedex, France
| | - Céline Mouly
- Service d'endocrinologie, maladies métaboliques et nutrition, pôle cardio-vasculaire et métabolique, CHU Larrey, 31059 Toulouse cedex, France
| | | | - Kathy Wagner
- Department of Pediatrics, CHU-Lenval, 06200 Nice, France
| | - Eugénie Koumakis
- Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Reference Center for Rare Genetic Bone Disorders, OSCAR Filière, Rheumatology Department, Cochin Hospital, AP-HP Centre-Paris University, INSERM U1160, Institut Imagine, 75014 Paris, France
| | - Christine Cortet-Rudelli
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
| | - Émilie Merlen
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
| | - Arnaud Jannin
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
- University of Lille, 59000 Lille, France
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer-Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Stéphanie Espiard
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
- University of Lille, 59000 Lille, France
| | - Isabelle Morange
- Department of Endocrinology, Aix Marseille Univ, APHM, INSERM, UMR1251 MMG, MARMARA Institute, CRMR HYPO, Hôpital de la Conception, 13005 Marseille, France
| | - Éric Baudin
- Department of Endocrine Oncology and Imaging, Gustave Roussy Cancer Campus Grand, 94800 Villejuif, France
| | - Mathias Cavaille
- U1240 Imagerie Moléculaire et Stratégies Théranostiques, INSERM, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, 63000 Clermont Ferrand, France
| | - Igor Tauveron
- Service d'Endocrinologie, Diabétologie et Maladies Métaboliques, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
- Laboratoire GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Marie-Pierre Teissier
- Unité INSERM 1094 & IRD, Université de Limoges, 87025 Limoges, France
- Service d'Endocrinologie-Diabétologie et Maladies métaboliques, Centre hospitalier universitaire Dupuytren 2, 87042 Limoges, France
| | - Françoise Borson-Chazot
- Hospices Civils de Lyon, Fédération d'Endocrinologie, Université Claude Bernard Lyon 1, 69003 Lyon, France
| | - Delphine Mirebeau-Prunier
- Unité Mixte de Recherche (UMR) MITOVASC, Laboratoire de Biochimie et Biologie Moléculaire, INSERM U1083, CNRS 6015, Université d'Angers, Centre Hospitalier Universitaire d'Angers, Angers 49933, France
| | - Frédérique Savagner
- Laboratory of Biochemistry and Molecular Biology, IFB-CHU, 31000 Toulouse, France
| | - Éric Pasmant
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, 75014 Paris, France
- Institut Cochin, Cancer Department, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, 75014 Paris, France
| | - Sophie Giraud
- Genetics Department, Hospices Civils de LYON (HCL), University Hospital, East Pathology Center, 69029 Bron Cedex, France
| | - Marie-Christine Vantyghem
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
- University of Lille, 59000 Lille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), European Genomic Institute for Diabetes (EGID), CHU Lille, 59000 Lille, France
| | - Pierre Goudet
- Service de Chirurgie Viscérale et Endocrinienne, Centre Hospitalier Universitaire François Mitterand, 21000 Dijon, France
| | - Anne Barlier
- Laboratory of Molecular Biology GEnOPé, Biogénopôle, Aix Marseille Univ, APHM, INSERM, UMR1251 MMG, Hôpital de la Timone, 13005 Marseille, France
| | - Catherine Cardot-Bauters
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, 59000 Lille, France
| | - Marie Françoise Odou
- CHU Lille, Service de Biochimie et Biologie moléculaire « Hormonologie, Métabolisme-Nutrition, Oncologie, 59000 Lille, France
- University of Lille, Inserm, CHU Lille, U1286-Infinite-Institute for Translational Research in Inflammation, 59000 Lille, France
| |
Collapse
|
8
|
Augsburger P, Liimatta J, Flück CE. Update on Adrenarche-Still a Mystery. J Clin Endocrinol Metab 2024; 109:1403-1422. [PMID: 38181424 DOI: 10.1210/clinem/dgae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
CONTEXT Adrenarche marks the timepoint of human adrenal development when the cortex starts secreting androgens in increasing amounts, in healthy children at age 8-9 years, with premature adrenarche (PA) earlier. Because the molecular regulation and significance of adrenarche are unknown, this prepubertal event is characterized descriptively, and PA is a diagnosis by exclusion with unclear long-term consequences. EVIDENCE ACQUISITION We searched the literature of the past 5 years, including original articles, reviews, and meta-analyses from PubMed, ScienceDirect, Web of Science, Embase, and Scopus, using search terms adrenarche, pubarche, DHEAS, steroidogenesis, adrenal, and zona reticularis. EVIDENCE SYNTHESIS Numerous studies addressed different topics of adrenarche and PA. Although basic studies on human adrenal development, zonation, and zona reticularis function enhanced our knowledge, the exact mechanism leading to adrenarche remains unsolved. Many regulators seem involved. A promising marker of adrenarche (11-ketotestosterone) was found in the 11-oxy androgen pathway. By current definition, the prevalence of PA can be as high as 9% to 23% in girls and 2% to 10% in boys, but only a subset of these children might face related adverse health outcomes. CONCLUSION New criteria for defining adrenarche and PA are needed to identify children at risk for later disease and to spare children with a normal variation. Further research is therefore required to understand adrenarche. Prospective, long-term studies should characterize prenatal or early postnatal developmental pathways that modulate trajectories of birth size, early postnatal growth, childhood overweight/obesity, adrenarche and puberty onset, and lead to abnormal sexual maturation, fertility, and other adverse outcomes.
Collapse
Affiliation(s)
- Philipp Augsburger
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Jani Liimatta
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland and Kuopio University Hospital, 70029 Kuopio, Finland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
9
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- 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, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
10
|
Sahakian N, Castinetti F, Romanet P, Reznik Y, Brue T. Updates on the genetics of multiple endocrine neoplasia. ANNALES D'ENDOCRINOLOGIE 2024; 85:127-135. [PMID: 38325596 DOI: 10.1016/j.ando.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 02/09/2024]
Abstract
Multiple endocrine neoplasia (MEN) is a group of syndromes with a genetic predisposition to the appearance of endocrine tumors, and shows autosomal dominant transmission. The advent of molecular genetics has led to improvements in the management of MEN in terms of diagnosis, prognosis and therapy. The genetics of MEN is the subject of regular updates, which will be presented throughout this paper. MEN1, the first to be described, is associated with the MEN1 gene. MEN1 is well known in terms of the observed phenotype, with genetic analysis being conclusive in 90% of patients with a typical phenotype, but is negative in around 10% of families with MEN1. Improvement in analysis techniques and the identification of other genes responsable for phenocopies allows the resolution of some, but not all, cases, notably non-familial forms suspected to be fortuitous assocations with tumors. MEN4 is a rare phenocopy of MEN1 linked to constitutional mutations in the CDKN1B gene. Though it closely resembles the phenotype of MEN1, published data suggests the appearance of tumors is later and less frequent in MEN4. MEN2, which results from mutations in the RET oncogene, shows a strong genotype-phenotype correlation. This correlation is particularly evident in the major manifestation of MEN2, medullary thyroid carcinoma (MTC), in which disease aggressiveness is dependent on the pathogenic variant of RET. However, recent studies cast doubt on this correlation between MTC and pathogenic variant. Lastly, the recent description of families carrying a mutation in MAX, which is known to predispose to the development of pheochromocytoma and paraganglioma, and presents a phenotypic spectrum that evokes MEN, suggests the existence of another syndrome, MEN5.
Collapse
Affiliation(s)
- Nicolas Sahakian
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France.
| | - Frederic Castinetti
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France
| | - Pauline Romanet
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Laboratory of Molecular Biology, Biogenopole, Timone University Hospital, AP-HM, Marseille, France
| | - Yves Reznik
- Department of endocrinology, diabetes, metabolic disorders, University Hospital Caen, Caen, France
| | - Thierry Brue
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France
| |
Collapse
|
11
|
Sada V, Puliani G, Feola T, Pirchio R, Pofi R, Sesti F, De Alcubierre D, Amodeo ME, D'Aniello F, Vincenzi L, Gianfrilli D, Isidori AM, Grossman AB, Sbardella E. Tall stature and gigantism in transition age: clinical and genetic aspects-a literature review and recommendations. J Endocrinol Invest 2024; 47:777-793. [PMID: 37891382 DOI: 10.1007/s40618-023-02223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE Tall stature is defined as height greater than the threshold of more than 2 standard deviations above the average population height for age, sex, and ethnicity. Many studies have described the main aspects of this condition during puberty, but an analysis of the characteristics that the physician should consider in the differential diagnosis of gigantism-tall stature secondary to a pituitary tumour-during the transition age (15-25 years) is still lacking. METHODS A comprehensive search of English-language original articles was conducted in the MEDLINE database (December 2021-March 2022). We selected all studies regarding epidemiology, genetic aspects, and the diagnosis of tall stature and gigantism during the transition age. RESULTS Generally, referrals for tall stature are not as frequent as expected because most cases are familial and are usually unreported by parents and patients to endocrinologists. For this reason, lacking such experience of tall stature, familiarity with many rarer overgrowth syndromes is essential. In the transition age, it is important but challenging to distinguish adolescents with high constitutional stature from those with gigantism. Pituitary gigantism is a rare disease in the transition age, but its systemic complications are very relevant for future health. Endocrine evaluation is crucial for identifying conditions that require hormonal treatment so that they can be treated early to improve the quality of life and prevent comorbidities of individual patient in this age range. CONCLUSION The aim of our review is to provide a practical clinical approach to recognise adolescents, potentially affected by gigantism, as early as possible.
Collapse
Affiliation(s)
- V Sada
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - G Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - T Feola
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - R Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - R Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford University Hospitals, NHS Trust, Oxford, UK
| | - F Sesti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - M E Amodeo
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - F D'Aniello
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - L Vincenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Centre for Rare Diseases (ENDO-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - A B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
| | - E Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
12
|
Einarsson HB, Frederiksen AL, Pedersen IS, Ettrup MS, Wirenfeldt M, Boldt H, Nguyen N, Andersen MS, Bjarkam CR, Poulsen FR. PDP type brain tumor in association with multiple endocrine neoplasia type 1. Heliyon 2024; 10:e27418. [PMID: 38510015 PMCID: PMC10951523 DOI: 10.1016/j.heliyon.2024.e27418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a rare autosomal dominant syndrome caused by inactivating pathogenic variants in the tumor suppressor gene menin 1 on chromosome 11q13 (Falchetti et al., 2009). The syndrome is characterized by neoplasia in two or more endocrine glands and has a high degree of penetrance. Pathogenic germline multiple neoplasia type 1 variants primarily result in neoplasia affecting the parathyroid glands, the pancreatic islet cells, and the anterior pituitary in combination. Primary hyperparathyroidism is the most common pathological manifestation of the syndrome, followed by pancreatic neuroendocrine tumors. Important genetic confirmation has been provided showing that ependymoma should be considered as a neoplasm that can occur in patients with MEN1 (Kato et al., 1996; Cuevas-Ocampo et al., 2017). The biphasic histopathological tumor entity shown in the present case we name Pleomorphic Xanthoastocytoma grade 3 differential pathology (PDP) in association with Multiple Endocrine Neoplasia type 1. This MEN1 associated tumor subtype is an extension of the findings on MEN1 associated ependymoma, where we show that the clinical phenotype itself may potentially be triggered by a frameshift germline pathogenic variant for the MEN1 gene, in combination with cyclin-dependent kinase inhibitor 1B gene germline variant and cyclin dependent kinase inhibitor 2A somatic deletion downstream of menin.
Collapse
Affiliation(s)
| | - Anja Lisbeth Frederiksen
- Molecular Diagnostics, Aalborg University Hospital and Clinical Cancer Research Center, Aalborg University Hospital, Denmark
- Department of Clinical Medicine, Aalborg University, Denmark
| | - Inge Soekilde Pedersen
- Molecular Diagnostics, Aalborg University Hospital and Clinical Cancer Research Center, Aalborg University Hospital, Denmark
- Department of Clinical Medicine, Aalborg University, Denmark
| | | | - Martin Wirenfeldt
- Department of Pathology, Hospital South West Jutland, Denmark
- Department of Regional Health Research, University of Southern, Denmark
- Department of Clinical Research and BRIDGE, Brain Research – Inter-Disciplinary Guided Excellence, University of Southern, Denmark
| | - Henning Boldt
- Department of Pathology, Odense University Hospital, Denmark
| | - Nina Nguyen
- Department of Neuroradiology, Odense University Hospital, Denmark
| | | | | | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital, Denmark
- Department of Clinical Research and BRIDGE, Brain Research – Inter-Disciplinary Guided Excellence, University of Southern, Denmark
| |
Collapse
|
13
|
De Leo A, Ruscelli M, Maloberti T, Coluccelli S, Repaci A, de Biase D, Tallini G. Molecular pathology of endocrine gland tumors: genetic alterations and clinicopathologic relevance. Virchows Arch 2024; 484:289-319. [PMID: 38108848 PMCID: PMC10948534 DOI: 10.1007/s00428-023-03713-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 12/19/2023]
Abstract
Tumors of the endocrine glands are common. Knowledge of their molecular pathology has greatly advanced in the recent past. This review covers the main molecular alterations of tumors of the anterior pituitary, thyroid and parathyroid glands, adrenal cortex, and adrenal medulla and paraganglia. All endocrine gland tumors enjoy a robust correlation between genotype and phenotype. High-throughput molecular analysis demonstrates that endocrine gland tumors can be grouped into molecular groups that are relevant from both pathologic and clinical point of views. In this review, genetic alterations have been discussed and tabulated with respect to their molecular pathogenetic role and clinicopathologic implications, addressing the use of molecular biomarkers for the purpose of diagnosis and prognosis and predicting response to molecular therapy. Hereditary conditions that play a key role in determining predisposition to many types of endocrine tumors are also discussed.
Collapse
Affiliation(s)
- Antonio De Leo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Martina Ruscelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
| | - Thais Maloberti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Sara Coluccelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Andrea Repaci
- Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, 40126, Bologna, Italy
| | - Giovanni Tallini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy.
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy.
| |
Collapse
|
14
|
Ruggeri RM, Benevento E, De Cicco F, Grossrubatscher EM, Hasballa I, Tarsitano MG, Centello R, Isidori AM, Colao A, Pellegata NS, Faggiano A. Multiple endocrine neoplasia type 4 (MEN4): a thorough update on the latest and least known men syndrome. Endocrine 2023; 82:480-490. [PMID: 37632635 DOI: 10.1007/s12020-023-03497-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE Multiple endocrine neoplasia type 4 (MEN4) is a rare multiglandular endocrine neoplasia syndrome, associated with a wide tumor spectrum but hallmarked by primary hyperparathyroidism, which represents the most common clinical feature, followed by pituitary (functional and non-functional) adenomas, and neuroendocrine tumors. MEN4 clinically overlaps MEN type 1 (MEN1) but differs from it for milder clinical features and an older patient's age at onset. The underlying mutated gene, CDKN1B, encodes the cell cycle regulator p27, implicated in cellular proliferation, motility and apoptosis. Given the paucity of MEN4 cases described in the literature, possible genotype-phenotype correlations have not been thoroughly assessed, and specific clinical recommendations are lacking. The present review provides an extensive overview of molecular genetics and clinical features of MEN4, with the aim of contributing to delineate peculiar strategies for clinical management, screening and follow-up of the last and least known MEN syndrome. METHODS A literature search was performed through online databases like MEDLINE and Scopus. CONCLUSIONS MEN4 is much less common that MEN1, tend to present later in life with a more indolent course, although involving the same primary organs as MEN1. As a consequence, MEN4 patients might need specific diagnostic and therapeutic approaches and a different strategy for screening and follow-up. Further studies are needed to assess the real oncological risk of MEN4 carriers, and to establish a standardized screening protocol. Furthermore, a deeper understanding of molecular genetics of MEN4 is needed in order to explore p27 as a novel therapeutic target.
Collapse
Affiliation(s)
- Rosaria M Ruggeri
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, 98125, Messina, Italy.
| | - Elio Benevento
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | | | | | - Iderina Hasballa
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
| | | | - Roberta Centello
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", Federico II University, Naples, Italy
| | | | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of excellence, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
15
|
Лапшина АМ. [Molecular genetic abnormalities in ACTH-secreting pituitary tumors (corticotropinomas): fundamental research and prospects for use in clinical practice]. PROBLEMY ENDOKRINOLOGII 2023; 70:23-30. [PMID: 39069770 PMCID: PMC11334230 DOI: 10.14341/probl13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/16/2023] [Accepted: 11/02/2023] [Indexed: 07/30/2024]
Abstract
In recent years, a large number of studies have been carried out to research molecular genetic abnormalities in ACTH--secreting pituitary tumors. This review presents a comprehensive analysis of exome studies results (germline and somatic mutations, chromosomal abnormalities in corticotropinomas which developed as part of hereditary syndromes MEN 1, 2, 4, DICER1, Carney complex etc., and isolated tumors, respectively) and transcriptome (specific genes expression profiles in hormonally active and inactive corticotropinomas, regulation of cell cycles and signal pathways). Modern technologies (next-generation sequencing - NGS) allow us to study the state of the microRNAome, DNA methylome and inactive chromatin sites, in particular using RNA sequencing. Thus, a wide range of fundamental studies is shown, the results of which allow us to identify and comprehend the key previously known and new pathogenesis mechanisms and biomarkers of corticotropinomas. The characteristics of the most promising molecular genetic factors that can be used in clinical practice for screening and earlier diagnosis of hereditary syndromes and isolated corticotropinomas, differential diagnosis of various forms of endogenous hypercorticism, sensitivity to existing and potential therapies and personalized outcome determination of Cushing`s disease.
Collapse
Affiliation(s)
- А. М. Лапшина
- Национальный медицинский исследовательский центр эндокринологии
| |
Collapse
|
16
|
Mazarico-Altisent I, Capel I, Baena N, Bella-Cueto MR, Barcons S, Guirao X, Pareja R, Muntean A, Arsentales V, Caixàs A, Rigla M. Genetic testing for familial hyperparathyroidism: clinical-genetic profile in a Mediterranean cohort. Front Endocrinol (Lausanne) 2023; 14:1244361. [PMID: 37810884 PMCID: PMC10558207 DOI: 10.3389/fendo.2023.1244361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Background Approximately 10% of primary hyperparathyroidism cases are hereditary, due to germline mutations in certain genes. Although clinically relevant, a systematized genetic diagnosis is missing due to a lack of firm evidence regarding individuals to test and which genes to evaluate. Methods A customized gene panel (AIP, AP2S1, CASR, CDC73, CDKN1A, CDKN1B, CDKN2B, CDKN2C, GCM2, GNA11, MEN1, PTH, RET, and TRPV6) was performed in 40 patients from the Mediterranean area with suspected familial hyperparathyroidism (≤45 years of age, family history, high-risk histology, associated tumour, multiglandular disease, or recurrent hyperparathyroidism). We aimed to determine the prevalence of germline variants in these patients, to clinically characterize the probands and their relatives, and to compare disease severity in carriers versus those with a negative genetic test. Results Germline variants were observed in 9/40 patients (22.5%): 2 previously unknown pathogenic/likely pathogenic variants of CDKN1B (related to MEN4), 1 novel variant of uncertain significance of CDKN2C, 4 variants of CASR (3 pathogenic/likely pathogenic variants and 1 variant of uncertain significance), and 2 novel variants of uncertain significance of TRPV6. Familial segregation studies allowed diagnosis and early treatment of PHPT in first-degree relatives of probands. Conclusion The observed prevalence of germline variants in the Mediterranean cohort under study was remarkable and slightly higher than that seen in other populations. Genetic screening for suspected familial hyperparathyroidism allows the early diagnosis and treatment of PHPT and other related comorbidities. We recommend genetic testing for patients with primary hyperparathyroidism who present with high-risk features.
Collapse
Affiliation(s)
- Isabel Mazarico-Altisent
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Ismael Capel
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Neus Baena
- Genetic Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Maria Rosa Bella-Cueto
- Pathology Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Santi Barcons
- Surgery Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Xavier Guirao
- Surgery Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Rocío Pareja
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Andreea Muntean
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Valeria Arsentales
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Assumpta Caixàs
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| | - Mercedes Rigla
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d’Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Sabadell, Barcelona, Spain
| |
Collapse
|
17
|
Faggiano A, Fazzalari B, Mikovic N, Russo F, Zamponi V, Mazzilli R, Guarnieri V, Piane M, Visco V, Petrucci S. Clinical Factors Predicting Multiple Endocrine Neoplasia Type 1 and Type 4 in Patients with Neuroendocrine Tumors. Genes (Basel) 2023; 14:1782. [PMID: 37761922 PMCID: PMC10531237 DOI: 10.3390/genes14091782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this study is to evaluate the predictive role of specific clinical factors for the diagnosis of Multiple Endocrine Neoplasia type-1 (MEN1) and type-4 (MEN4) in patients with an initial diagnosis of gastrointestinal, bronchial, or thymic neuroendocrine tumor (NET). METHODS Patients referred to the NET Unit between June 2021 and December 2022 with a diagnosis of NET and at least one clinical criterion of suspicion for MEN1 and MEN4 underwent molecular analysis of the MEN1 and CDKN1B genes. Phenotypic criteria were: (1) age ≤ 40 years; (2) NET multifocality; (3) MEN1/4-associated manifestations other than NETs; and (4) endocrine syndrome related to NETs or pituitary/adrenal tumors. RESULTS A total of 22 patients were studied. In 18 patients (81.8%), the first-level genetic test was negative (Group A), while four patients (25%) were positive for MEN1 (Group B). No patient was positive for MEN4. In Group A, 10 cases had only one clinical criterion, and three patients met three criteria. In Group B, three patients had three criteria, and one met all criteria. CONCLUSION These preliminary data show that a diagnosis of NET in patients with a negative family history is suggestive of MEN1 in the presence of ≥three positive phenotypic criteria, including early age, multifocality, multiple MEN-associated manifestations, and endocrine syndromes. This indication may allow optimization of the diagnosis of MEN in patients with NET.
Collapse
Affiliation(s)
- Antongiulio Faggiano
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Beatrice Fazzalari
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Nevena Mikovic
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Flaminia Russo
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Virginia Zamponi
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Rossella Mazzilli
- Endocrinology Unit, Sant’Andrea Hospital, ENETS Center of Excellence, 00189 Rome, Italy; (B.F.); (N.M.); (F.R.); (V.Z.); (R.M.)
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
| | - Vito Guarnieri
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 Foggia, Italy;
| | - Maria Piane
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
- UOD Medical Genetics and Advanced Cell Diagnostics, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Vincenzo Visco
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
- UOD Medical Genetics and Advanced Cell Diagnostics, Sant’Andrea Hospital, 00189 Rome, Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (M.P.); (V.V.); (S.P.)
- UOD Medical Genetics and Advanced Cell Diagnostics, Sant’Andrea Hospital, 00189 Rome, Italy
| |
Collapse
|
18
|
Vamvoukaki R, Chrysoulaki M, Betsi G, Xekouki P. Pituitary Tumorigenesis-Implications for Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040812. [PMID: 37109772 PMCID: PMC10145673 DOI: 10.3390/medicina59040812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs), the third most common intracranial tumor, are mostly benign. However, some of them may display a more aggressive behavior, invading into the surrounding structures. While they may rarely metastasize, they may resist different treatment modalities. Several major advances in molecular biology in the past few years led to the discovery of the possible mechanisms involved in pituitary tumorigenesis with a possible therapeutic implication. The mutations in the different proteins involved in the Gsa/protein kinase A/c AMP signaling pathway are well-known and are responsible for many PitNETS, such as somatotropinomas and, in the context of syndromes, as the McCune-Albright syndrome, Carney complex, familiar isolated pituitary adenoma (FIPA), and X-linked acrogigantism (XLAG). The other pathways involved are the MAPK/ERK, PI3K/Akt, Wnt, and the most recently studied HIPPO pathways. Moreover, the mutations in several other tumor suppressor genes, such as menin and CDKN1B, are responsible for the MEN1 and MEN4 syndromes and succinate dehydrogenase (SDHx) in the context of the 3PAs syndrome. Furthermore, the pituitary stem cells and miRNAs hold an essential role in pituitary tumorigenesis and may represent new molecular targets for their diagnosis and treatment. This review aims to summarize the different cell signaling pathways and genes involved in pituitary tumorigenesis in an attempt to clarify their implications for diagnosis and management.
Collapse
Affiliation(s)
- Rodanthi Vamvoukaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Maria Chrysoulaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Grigoria Betsi
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| |
Collapse
|
19
|
Mazarico-Altisent I, Capel I, Baena N, Bella-Cueto MR, Barcons S, Guirao X, Albert L, Cano A, Pareja R, Caixàs A, Rigla M. Novel germline variants of CDKN1B and CDKN2C identified during screening for familial primary hyperparathyroidism. J Endocrinol Invest 2023; 46:829-840. [PMID: 36334246 PMCID: PMC10023768 DOI: 10.1007/s40618-022-01948-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE CDKN1B mutations were established as a cause of multiple endocrine neoplasia 4 (MEN4) syndrome in patients with MEN1 phenotype without a mutation in the MEN1 gene. In addition, variants in other cyclin-dependent kinase inhibitors (CDKIs) were found in some MEN1-like cases without the MEN1 mutation. We aimed to describe novel germline mutations of these genes in patients with primary hyperparathyroidism (PHPT). METHODS During genetic screening for familial hyperparathyroidism, three novel CDKIs germline mutations in three unrelated cases between January 2019 and November 2021 were identified. In this report, we describe clinical features, DNA sequence analysis, and familial segregation studies based on these patients and their relatives. Genome-wide DNA study of loss of heterozygosity (LOH), copy number variation (CNV), and p27/kip immunohistochemistry was performed on tumour samples. RESULTS DNA screening was performed for atypical parathyroid adenomas in cases 1 and 2 and for cystic parathyroid adenoma and young age at diagnosis of PHPT in case 3. Genetic analysis identified likely pathogenic variants of CDKN1B in cases 1 and 2 and a variant of the uncertain significance of CDKN2C, with uniparental disomy in the tumour sample, in case 3. Neoplasm screening of probands showed other non-endocrine tumours in case 1 (colon adenoma with dysplasia and atypical lipomas) and case 2 (aberrant T-cell population) and a non-functional pituitary adenoma in case 3. CONCLUSION Germline mutations in CDKIs should be included in gene panels for genetic testing of primary hyperparathyroidism. New germline variants here described can be added to the current knowledge.
Collapse
Affiliation(s)
- I Mazarico-Altisent
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain.
| | - I Capel
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - N Baena
- Genetic Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - M R Bella-Cueto
- Pathology Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - S Barcons
- Surgery Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - X Guirao
- Surgery Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - L Albert
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - A Cano
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - R Pareja
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - A Caixàs
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - M Rigla
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| |
Collapse
|
20
|
Abstract
Hereditary pituitary tumorigenesis is seen in a relatively small proportion (around 5%) of patients with pituitary neuroendocrine tumors (PitNETs). The aim of the current review is to describe the main clinical and molecular features of such pituitary tumors associated with hereditary or familial characteristics, many of which have now been genetically identified. The genetic patterns of inheritance are classified into isolated familial PitNETs and the syndromic tumors. In general, the established genetic causes of familial tumorigenesis tend to present at a younger age, often pursue a more aggressive course, and are more frequently associated with growth hormone hypersecretion compared to sporadic tumors. The mostly studied molecular pathways implicated are the protein kinase A and phosphatidyl-inositol pathways, which are in the main related to mutations in the syndromes of familial isolated pituitary adenoma (FIPA), Carney complex syndrome, and X-linked acrogigantism. Another well-documented mechanism consists of the regulation of p27 or p21 proteins, with further acceleration of the pituitary cell cycle through the check points G1/S and M/G1, mostly documented in multiple endocrine neoplasia type 4. In conclusion, PitNETs may occur in relation to well-established familial germline mutations which may determine the clinical phenotype and the response to treatment, and may require family screening.
Collapse
Affiliation(s)
- Eleni Armeni
- Dept. of Endocrinology, Royal Free Hospital, London, NW3 2QG, UK.
| | - Ashley Grossman
- Dept. of Endocrinology, Royal Free Hospital, London, NW3 2QG, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- Green Templeton College, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Jacquart A, Schilter KF, Udani R, Evans D, Carroll TB, Reddi HV. Constitutional mosaicism for a pathogenic variant in MEN1 associated with multiple endocrine neoplasia type I syndrome. Clin Genet 2023. [PMID: 36840469 DOI: 10.1111/cge.14314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Affiliation(s)
- Amanda Jacquart
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Kala F Schilter
- Precision Medicine Laboratory, Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Rupa Udani
- Precision Medicine Laboratory, Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Douglas Evans
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ty B Carroll
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Honey V Reddi
- Precision Medicine Laboratory, Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
22
|
Singeisen H, Renzulli MM, Pavlicek V, Probst P, Hauswirth F, Muller MK, Adamczyk M, Weber A, Kaderli RM, Renzulli P. Multiple endocrine neoplasia type 4: a new member of the MEN family. Endocr Connect 2023; 12:e220411. [PMID: 36520683 PMCID: PMC9874964 DOI: 10.1530/ec-22-0411] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Multiple endocrine neoplasia type 4 (MEN4) is caused by a CDKN1B germline mutation first described in 2006. Its estimated prevalence is less than one per million. The aim of this study was to define the disease characteristics. METHODS A systematic review was performed according to the PRISMA 2020 criteria. A literature search from January 2006 to August 2022 was done using MEDLINE® and Web of ScienceTM. RESULTS Forty-eight symptomatic patients fulfilled the pre-defined eligibility criteria. Twenty-eight different CDKN1B variants, mostly missense (21/48, 44%) and frameshift mutations (17/48, 35%), were reported. The majority of patients were women (36/48, 75%). Men became symptomatic at a median age of 32.5 years (range 10-68, mean 33.7 ± 23), whereas the same event was recorded for women at a median age of 49.5 years (range 5-76, mean 44.8 ± 19.9) (P = 0.25). The most frequently affected endocrine organ was the parathyroid gland (36/48, 75%; uniglandular disease 31/36, 86%), followed by the pituitary gland (21/48, 44%; hormone-secreting 16/21, 76%), the endocrine pancreas (7/48, 15%), and the thyroid gland (4/48, 8%). Tumors of the adrenal glands and thymus were found in three and two patients, respectively. The presenting first endocrine pathology concerned the parathyroid (27/48, 56%) and the pituitary gland (11/48, 23%). There were one (27/48, 56%), two (13/48, 27%), three (3/48, 6%), or four (5/48, 10%) syn- or metachronously affected endocrine organs in a single patient, respectively. CONCLUSION MEN4 is an extremely rare disease, which most frequently affects women around 50 years of age. Primary hyperparathyroidism as a uniglandular disease is the leading pathology.
Collapse
Affiliation(s)
- Hélène Singeisen
- Department of Internal Medicine, Endocrinology, Cantonal Hospital Thurgau, Münsterlingen, Switzerland
| | | | - Vojtech Pavlicek
- Department of Internal Medicine, Endocrinology, Cantonal Hospital Thurgau, Münsterlingen, Switzerland
| | - Pascal Probst
- Department of Surgery, Cantonal Hospital Thurgau, Frauenfeld, Switzerland
| | - Fabian Hauswirth
- Department of Surgery, Cantonal Hospital Thurgau, Münsterlingen, Switzerland
| | - Markus K Muller
- Department of Surgery, Cantonal Hospital Thurgau, Frauenfeld, Switzerland
| | - Magdalene Adamczyk
- Department of Pathology and Molecular Pathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Reto Martin Kaderli
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Pietro Renzulli
- Department of Surgery, Cantonal Hospital Thurgau, Münsterlingen, Switzerland
- Correspondence should be addressed to P Renzulli:
| |
Collapse
|
23
|
Ruggeri RM, Benevento E, De Cicco F, Fazzalari B, Guadagno E, Hasballa I, Tarsitano MG, Isidori AM, Colao A, Faggiano A. Neuroendocrine neoplasms in the context of inherited tumor syndromes: a reappraisal focused on targeted therapies. J Endocrinol Invest 2023; 46:213-234. [PMID: 36038743 DOI: 10.1007/s40618-022-01905-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/16/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE Neuroendocrine neoplasms can occur as part of inherited disorders, usually in the form of well-differentiated, slow-growing tumors (NET). The main predisposing syndromes include: multiple endocrine neoplasias type 1 (MEN1), associated with a large spectrum of gastroenteropancreatic and thoracic NETs, and type 4 (MEN4), associated with a wide tumour spectrum similar to that of MEN1; von Hippel-Lindau syndrome (VHL), tuberous sclerosis (TSC), and neurofibromatosis 1 (NF-1), associated with pancreatic NETs. In the present review, we propose a reappraisal of the genetic basis and clinical features of gastroenteropancreatic and thoracic NETs in the setting of inherited syndromes with a special focus on molecularly targeted therapies for these lesions. METHODS Literature search was systematically performed through online databases, including MEDLINE (via PubMed), and Scopus using multiple keywords' combinations up to June 2022. RESULTS Somatostatin analogues (SSAs) remain the mainstay of systemic treatment for NETs, and radiolabelled SSAs can be used for peptide-receptor radionuclide therapy for somatostatin receptor (SSTR)-positive NETs. Apart of these SSTR-targeted therapies, other targeted agents have been approved for NETs: the mTOR inhibitor everolimus for lung, gastroenteropatic and unknown origin NET, and sunitinib, an antiangiogenic tyrosine kinase inhibitor, for pancreatic NET. Novel targeted therapies with other antiangiogenic agents and immunotherapies have been also under evaluation. CONCLUSIONS Major advances in the understanding of genetic and epigenetic mechanisms of NET development in the context of inherited endocrine disorders have led to the recognition of molecular targetable alterations, providing a rationale for the implementation of treatments and development of novel targeted therapies.
Collapse
Affiliation(s)
- R M Ruggeri
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Messina, AOU Policlinico "Gaetano Martino" University Hospital, 98125, Messina, Italy.
| | - E Benevento
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Naples, Italy
| | - F De Cicco
- SSD Endocrine Disease and Diabetology, ASL TO3, Pinerolo, TO, Italy
| | - B Fazzalari
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - E Guadagno
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Naples, Italy
| | - I Hasballa
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - M G Tarsitano
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - A M Isidori
- Gruppo NETTARE, Policlinico Umberto I, Università Sapienza, Rome, Italy
| | - A Colao
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", Federico II University, Naples, Italy
| | - A Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
24
|
Halperin R, Arnon L, Nasirov S, Friedensohn L, Gershinsky M, Telerman A, Friedman E, Bernstein-Molho R, Tirosh A. Germline CDKN1B variant type and site are associated with phenotype in MEN4. Endocr Relat Cancer 2023; 30:ERC-22-0174. [PMID: 36256846 DOI: 10.1530/erc-22-0174] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Multiple endocrine neoplasia 4 (MEN4) is a rare multiglandular endocrine neoplasia syndrome clinically hallmarked by primary hyperparathyroidism (PHPT), pituitary adenoma (PitAd), and neuroendocrine tumors (NET), clinically overlapping MEN1. The underlying mutated gene - CDKN1B, encodes for the cell-cycle regulator p27. Possible genotype-phenotype correlations in MEN4 have not been thoroughly assessed. Prompted by the findings in three Israeli MEN4 kindreds, we performed a literature review on published and unpublished data from previously reported MEN4/CDKN1B cases. Univariate analysis analyzed time-dependent risks for developing PHPT, PitAd, or NET by variant type and position along the gene. Overall, 74 MEN4 cases were analyzed. PHPT risk was 53.4% by age 60 years (mean age at diagnosis age 50.6 ± 13.9 years), risk for PitAd was 23.2% and risk for NET was 16.2% (34.4 ± 21.4 and 52.9 ± 13.9 years, respectively). The frameshift variant p.Q107fs was the most common variant identified (4/41 (9.7%) kindreds). Patients with indels had higher risk for PHPT vs point mutations (log-rank, P = 0.029). Variants in codons 94-96 were associated with higher risk for PHPT (P < 0.001) and PitAd (P = 0.031). To conclude, MEN4 is clinically distinct from MEN1, with lower risk and older age for PHPT diagnosis. We report recurrent CDKN1B frameshift variants and possible genotype-phenotype correlations.
Collapse
Affiliation(s)
- Reut Halperin
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Liat Arnon
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Sapir Nasirov
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| | - Limor Friedensohn
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Gershinsky
- Department of Endocrinology and Diabetes, Lady Davis Carmel Medical Center and Linn Medical Center and Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alona Telerman
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Eitan Friedman
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- Personalized Preventive Genetics Center, Assuta Medical Center, Tel-Aviv, Israel
| | - Rinat Bernstein-Molho
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
- The Susanne Levy Gertner Oncogenetics Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Amit Tirosh
- ENTIRE Endocrine Neoplasia Translational Research Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
25
|
Agrawal N, Gersey ZC, Abou-Al-Shaar H, Gardner PA, Mantica M, Agnihotri S, Mahmud H, Fazeli PK, Zenonos GA. Major Genetic Motifs in Pituitary Adenomas: A Practical Literature Update. World Neurosurg 2023; 169:43-50. [PMID: 36115566 PMCID: PMC11195535 DOI: 10.1016/j.wneu.2022.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND The literature includes many studies examining the genetic abnormalities that influence pituitary adenomas (PAs). We aimed to state the collective knowledge on the genetic underpinnings of PAs by organizing, summarizing, and consolidating the literature to serve as a comprehensive review for scientists and clinicians of the most up-to-date information underlying the genetic landscape of PAs. METHODS The PubMed and Google Scholar databases were searched using multiple key words and combined Medical Subject Headings terms; only articles published in English between January 2000 and January 2022 were included. Articles in which the focus did not relate to genetics, that included mainly anecdotal evidence, or that were single case studies were eliminated. RESULTS PAs are one of the most common intracranial neoplasms. However, the genetic underpinnings for these tumors are not yet fully elucidated. There are several categories of PAs: clinically significant versus not clinically significant, functional versus nonfunctional, and germline-derived versus sporadic origin. Each of these disease subcategories is characterized by unique genetic aberrations. Recently, more genes and other types of genetic aberrations have been identified as possible causes of PAs, such as copy number variations and altered levels of microRNAs. CONCLUSIONS This review serves to consolidate and summarize the literature discussing the genetic motifs of PAs to help physicians and scientists deliver patient-centered therapies.
Collapse
Affiliation(s)
- Nishant Agrawal
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zachary C Gersey
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussam Abou-Al-Shaar
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Paul A Gardner
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Megan Mantica
- Departments of Neuro-Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sameer Agnihotri
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussain Mahmud
- Departments of Endocrinology and Metabolism, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Pouneh K Fazeli
- Departments of Endocrinology and Metabolism, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Georgios A Zenonos
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
26
|
The Classic, the Trendy, and the Refashioned: A Primer for Pathologists on What Is New in Familial Endocrine Tumor Syndromes. Adv Anat Pathol 2023; 30:69-78. [PMID: 36136401 DOI: 10.1097/pap.0000000000000370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Familial endocrine tumor syndromes are continuously expanding owing to the growing role of genetic testing in routine clinical practice. Pathologists are usually the first on the clinical team to encounter these syndromes at their initial presentation; thus, recognizing them is becoming more pivotal in routine pathology practice to help in properly planning management and further family testing. Our increasing knowledge about them is reflected in the newer syndromes included in the new World Health Organization classification and in the evolving discovery of new endocrine tumors and new familial associations. In many of these syndromes, the clinical features and co-occurrence of multiple neoplasia are the only clues (multiple endocrine neoplasia syndromes). In other syndromes, specific morphologic findings (pituitary blastoma and DICER1 syndrome, cribriform morular thyroid carcinoma, and AFP syndrome) and available ancillary studies (SDHB in SDH-deficient tumor syndromes) can aid pathologists. The aim of this review is to provide a primer on recent updates on familial endocrine tumor syndromes and related tumors, focusing on recent classification changes or tumor syndromes where a clearer role for pathologists is at play.
Collapse
|
27
|
Abstract
Gonadotroph cell adenoma is the most common clinically nonfunctioning pituitary adenoma; and is pathologically defined by immunopositivity for SF-1, GATA2 and ER-α. Most tumors also stain for follicle stimulating hormone and luteinizing hormone β-subunits, but are usually hormonally silent and discovered incidentally or due to local mass effects. Complete transsphenoidal resection should be attempted when surgery is indicated. Post-surgical treatment can include radiation of the tumor remnant and medical treatment. Among medical treatments, dopamine agonists show the best evidence for preventing the need for recurrent surgery or radiation, and should be considered in patients with relatively bulky remnants or who have high risk features associated with tumor progression. Temozolomide is indicated for aggressive adenomas and carcinomas. Less well-established treatments include somatostatin receptor ligands, peptide receptor radionucleotide therapy and immunomodulatory agents.
Collapse
Affiliation(s)
- Naomi Even-Zohar
- Institute of Endocrinology, Diabetes, Metabolism, and Hypertension, Tel Aviv-Sourasky Medical Center, Israel.
| | - Yona Greenman
- Institute of Endocrinology, Diabetes, Metabolism, and Hypertension, Tel Aviv-Sourasky Medical Center, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel.
| |
Collapse
|
28
|
Coopmans EC, Korbonits M. Molecular genetic testing in the management of pituitary disease. Clin Endocrinol (Oxf) 2022; 97:424-435. [PMID: 35349723 DOI: 10.1111/cen.14706] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Most pituitary tumours occur sporadically without a genetically identifiable germline abnormality, a small but increasing proportion present with a genetic defect that predisposes to pituitary tumour development, either isolated (e.g., aryl hydrocarbon receptor-interacting protein, AIP) or as part of a tumour-predisposing syndrome (e.g., multiple endocrine neoplasia (MEN) type 1, Carney complex, McCune-Albright syndrome or pituitary tumour and paraganglioma association). Genetic alterations in sporadic pituitary adenomas may include somatic mutations (e.g., GNAS, USP8). In this review, we take a practical approach: which genetic syndromes should be considered in case of different presentation, such as tumour type, family history, age of onset and additional clinical features of the patient. DESIGN Review of the recent literature in the field of genetics of pituitary tumours. RESULTS Genetic testing in the management of pituitary disease is recommended in a significant minority of the cases. Understanding the genetic basis of the disease helps to identify patients and at-risk family members, facilitates early diagnosis and therefore better long-term outcome and opens up new pathways leading to tumorigenesis. CONCLUSION We provide a concise overview of the genetics of pituitary tumours and discuss the current challenges and implications of these genetic findings in clinical practice.
Collapse
Affiliation(s)
- Eva C Coopmans
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Medicine, Division of Endocrinology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Medicine, Endocrinology section, Pituitary Center Rotterdam, Erasmus University Medical Cente, Rotterdam, The Netherlands
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
29
|
Newey PJ, Hannan FM, Wilson A, Thakker RV. Genetics of monogenic disorders of calcium and bone metabolism. Clin Endocrinol (Oxf) 2022; 97:483-501. [PMID: 34935164 PMCID: PMC7614875 DOI: 10.1111/cen.14644] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/24/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022]
Abstract
Disorders of calcium homeostasis are the most frequent metabolic bone and mineral disease encountered by endocrinologists. These disorders usually manifest as primary hyperparathyroidism (PHPT) or hypoparathyroidism (HP), which have a monogenic aetiology in 5%-10% of cases, and may occur as an isolated endocrinopathy, or as part of a complex syndrome. The recognition and diagnosis of these disorders is important to facilitate the most appropriate management of the patient, with regard to both the calcium-related phenotype and any associated clinical features, and also to allow the identification of other family members who may be at risk of disease. Genetic testing forms an important tool in the investigation of PHPT and HP patients and is usually reserved for those deemed to be an increased risk of a monogenic disorder. However, identifying those suitable for testing requires a thorough clinical evaluation of the patient, as well as an understanding of the diversity of relevant phenotypes and their genetic basis. This review aims to provide an overview of the genetic basis of monogenic metabolic bone and mineral disorders, primarily focusing on those associated with abnormal calcium homeostasis, and aims to provide a practical guide to the implementation of genetic testing in the clinic.
Collapse
Affiliation(s)
- Paul J Newey
- Division of Molecular and Clinical Medicine, Ninewells Hospital & Medical School, University of Dundee, Scotland, UK
| | - Fadil M Hannan
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Abbie Wilson
- Division of Molecular and Clinical Medicine, Ninewells Hospital & Medical School, University of Dundee, Scotland, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism (OCDEM), Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Mohindroo C, McAllister F, De Jesus-Acosta A. Genetics of Pancreatic Neuroendocrine Tumors. Hematol Oncol Clin North Am 2022; 36:1033-1051. [PMID: 36154786 DOI: 10.1016/j.hoc.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pancreatic neuroendocrine tumors (pNETs) represent a relatively rare disease; however, the incidence has been increasing during the last 2 decades. Next generation sequencing has greatly increased our understanding of driver mutations in pNETs. Sporadic pNETs have consistently presented with mutations in MEN1, DAXX/ATRX, and genes related to the mammalian target of rapamycin pathway. Inherited pNETs have traditionally been associated with multiple endocrine neoplasia type 1, von Hippel-Lindau syndrome, neurofibromatosis type 1, and tuberous sclerosis complex. The current review expands on the existing knowledge and the relevant updates on the genetics of pNETs.
Collapse
Affiliation(s)
- Chirayu Mohindroo
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 1360, Houston, TX 77030, USA; Department of Internal Medicine, Sinai Hospital of Baltimore, 2435 W. Belvedere Ave, Ste 56, Baltimore, MD 21215, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 1360, Houston, TX 77030, USA; Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana De Jesus-Acosta
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, CRB1, 1650 Orleans Street, CRB1 Rm 409, Baltimore, MD 21287.
| |
Collapse
|
31
|
Yamamoto M, Takahashi Y. Genetic and Epigenetic Pathogenesis of Acromegaly. Cancers (Basel) 2022; 14:cancers14163861. [PMID: 36010855 PMCID: PMC9405703 DOI: 10.3390/cancers14163861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Various genetic and epigenetic factors are involved in the pathogenesis of somatotroph tumors. Although GNAS mutations are the most prevalent cause of somatotroph tumors, the cause of half of all pathogenesis occurrences remains unclarified. However, recent findings including the pangenomic analysis, such as genome, transcriptome, and methylome approaches, and histological characteristics of pituitary tumors, the involvement of AIP and GPR101, the mechanisms of genomic instability, and possible involvement of miRNAs have gradually unveiled the whole landscape of underlying mechanisms of somatotroph tumors. In this review, we will focus on the recent advances in the pathogenesis of somatotroph tumors. Abstract Acromegaly is caused by excessive secretion of GH and IGF-I mostly from somatotroph tumors. Various genetic and epigenetic factors are involved in the pathogenesis of somatotroph tumors. While somatic mutations of GNAS are the most prevalent cause of somatotroph tumors, germline mutations in various genes (AIP, PRKAR1A, GPR101, GNAS, MEN1, CDKN1B, SDHx, MAX) are also known as the cause of somatotroph tumors. Moreover, recent findings based on multiple perspectives of the pangenomic approach including genome, transcriptome, and methylome analyses, histological characterization, genomic instability, and possible involvement of miRNAs have gradually unveiled the whole landscape of the underlying mechanisms of somatotroph tumors. In this review, we will focus on the recent advances in genetic and epigenetic pathogenesis of somatotroph tumors.
Collapse
Affiliation(s)
- Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Correspondence: ; Tel.: +81-78-382-5861
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Department of Diabetes and Endocrinology, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
32
|
Simon J, Theodoropoulou M. Genetics of Cushing's disease. J Neuroendocrinol 2022; 34:e13148. [PMID: 35596671 DOI: 10.1111/jne.13148] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022]
Abstract
Corticotroph tumours are primarily sporadic monoclonal neoplasms and only rarely found in genetic syndromes. Recurrent mutations in the ubiquitin specific protease 8 (USP8) gene are found in around half of cases. Mutations in other genes such as USP48 and NR3C1 are less frequent, found in less than ~20% of cases. TP53 and ATXR mutations are reported in up to one out of four cases, when focusing in USP8 wild type or aggressive corticotroph tumours and carcinomas. At present, USP8 mutations are the primary driver alterations in sporadic corticotroph tumours, TP53 and ATXR mutations may indicate transition to more aggressive tumour phenotype. Next generation sequencing efforts have identified additional genomic alterations, whose role and importance in corticotroph tumorigenesis remains to be elucidated.
Collapse
Affiliation(s)
- Julia Simon
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
33
|
Seabrook A, Wijewardene A, De Sousa S, Wong T, Sheriff N, Gill AJ, Iyer R, Field M, Luxford C, Clifton-Bligh R, McCormack A, Tucker K. MEN4, the MEN1 Mimicker: A Case Series of three Phenotypically Heterogenous Patients With Unique CDKN1B Mutations. J Clin Endocrinol Metab 2022; 107:2339-2349. [PMID: 35323929 PMCID: PMC9282358 DOI: 10.1210/clinem/dgac162] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Indexed: 12/29/2022]
Abstract
CONTEXT Germline CDKN1B pathogenic variants result in multiple endocrine neoplasia type 4 (MEN4), an autosomal dominant hereditary tumor syndrome variably associated with primary hyperparathyroidism, pituitary adenoma, and duodenopancreatic neuroendocrine tumors. OBJECTIVE To report the phenotype of 3 unrelated cases each with a unique germline CDKN1B variant (of which 2 are novel) and compare these cases with those described in the current literature. DESIGN/METHODS Three case studies, including clinical presentation, germline, and tumor genetic analysis and family history. SETTING Two tertiary University Hospitals in Sydney, New South Wales, and 1 tertiary University Hospital in Canberra, Australian Capital Territory, Australia. OUTCOME Phenotype of the 3 cases and their kindred; molecular analysis and tumor p27kip1 immunohistochemistry. RESULTS Family A: The proband developed multiglandular primary hyperparathyroidism, a microprolactinoma and a multifocal nonfunctioning duodenopancreatic neuroendocrine tumor. Family B: The proband was diagnosed with primary hyperparathyroidism from a single parathyroid adenoma. Family C: The proband was diagnosed with a nonfunctioning pituitary microadenoma and ectopic Cushing's syndrome from an atypical thymic carcinoid tumor. Germline sequencing in each patient identified a unique variant in CDKN1B, 2 of which are novel (c.179G > A, p.Trp60*; c.475G > A, p.Asp159Asn) and 1 previously reported (c.374_375delCT, p.Ser125*). CONCLUSIONS Germline CDKN1B pathogenic variants cause the syndrome MEN4. The phenotype resulting from the 3 pathogenic variants described in this series highlights the heterogenous nature of this syndrome, ranging from isolated primary hyperparathyroidism to the full spectrum of endocrine manifestations. We report the first described cases of a prolactinoma and an atypical thymic carcinoid tumor in MEN4.
Collapse
Affiliation(s)
- Amanda Seabrook
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ayanthi Wijewardene
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sunita De Sousa
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, 5000
- South Australian Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Tang Wong
- The University of New South Wales, Sydney, NSW, 2052, Australia
- The University of Western Sydney, Sydney, NSW, 2560, Australia
- Department of Endocrinology, Prince of Wales Hospital, Sydney, NSW, 2064, Australia
| | - Nisa Sheriff
- Department of Endocrinology, Hornsby Ku-ring-gai Hospital, Sydney, NSW, 2077, Australia
| | - Anthony J Gill
- The University of Sydney, Sydney, NSW, 2006, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, 2064, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2064, Australia
| | - Rakesh Iyer
- Calvary Public Hospital, Canberra, ACT, 2617, Australia
| | - Michael Field
- Familial Cancer Service, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Catherine Luxford
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | | | | | - Katherine Tucker
- Correspondence: Katherine Tucker, MBBS, FRACP, AO, Hereditary Cancer Service Nelune Comprehensive Cancer Centre (Bright Building), 64-66 High St, Randwick, NSW, 2031, Australia.
| |
Collapse
|
34
|
Watanabe A, Wiseman SM. Multiple endocrine neoplasia type 4 & primary hyperparathyroidism: What the surgeon needs to know. Am J Surg 2022; 224:1017-1018. [DOI: 10.1016/j.amjsurg.2022.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022]
|
35
|
Pancreatic Neuroendocrine Neoplasms: Updates on Genomic Changes in Inherited Tumour Syndromes and Sporadic Tumours Based on WHO Classification. Crit Rev Oncol Hematol 2022; 172:103648. [PMID: 35248713 DOI: 10.1016/j.critrevonc.2022.103648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 02/19/2022] [Accepted: 02/28/2022] [Indexed: 12/16/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are the neuroendocrine neoplasms with greatest rate of increase in incidence. Approximately 10% of PanNENs arise as inherited tumour syndromes which include multiple endocrine neoplasia type 1, multiple endocrine neoplasia type 4, von Hippel-Lindau syndrome, neurofibromatosis type1, tuberous sclerosis complex 1/2, Cowden syndrome, and Glucagon cell hyperplasia and neoplasia as well as familial insulinomatosis. In sporadic PanNENs, driver mutations in MEN1, DAXX/ATRX and mTOR pathway genes are associated with development and progression in pancreatic neuroendocrine tumours. The other changes are in VEGF pathway, Notch pathway, germline mutations in MUTYH, CHEK2, BRCA2, PHLDA3 as well as other genetic alterations. On the other hand, pancreatic neuroendocrine carcinomas share similar genetic alterations with ductal adenocarcinomas, e.g., TP53, RB1 or KRAS. In addition, microRNA and changes in immune microenvironment were noted in PanNENs. Updates on these genetic knowledges contribute to the development of management strategies for patients with PanNENs.
Collapse
|
36
|
Damianse SSP, Nascimento GC, Rocha VCC, Nascimento AGPAC, Gaido NC, Azulay RSS, Dos Santos WC, Rodrigues VP, Quidute ARP, Magalhães M, Faria MDS. The Importance of Periodical Screening for Primary Hyperparathyroidism in a Pituitary Tumor Cohort in Searching Patients with MEN1 and its Genetic Profile. Endocr Pract 2022; 28:509-514. [PMID: 35247589 DOI: 10.1016/j.eprac.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Multiple endocrine neoplasia type 1 (MEN1) is a rare genetic syndrome characterized by parathyroid, anterior pituitary, and/or duodenopancreatic neuroendocrine tumors. Studies have indicated that investigating primary hyperparathyroidism (pHPT) with subsequent genetic screening may be an essential tool for the early diagnosis of MEN1 in patients with pituitary tumors (PTs). This study aimed to investigate the presence of pHPT in patients with PTs and, subsequently, to screen for genetic mutations and related tumors in those with MEN1 Syndrome. METHODS This study included 255 patients with PTs who were assessed for the presence of MEN1 from serum calcium and parathyroid hormone measurements. Mutation screening of the MEN1, CDKN1B, and AIP genes was performed in the index cases showing the MEN1 phenotype. RESULTS Five PT-evaluated patients presented a clinical condition compatible with MEN1. These patients had a younger age of onset and a more severe clinical condition. Genetic analysis identified a frameshift mutation in the MEN1 gene in one of the cases with the MEN1 phenotype, but point mutations in CDKN1B and AIP were not detected in any of these patients. CONCLUSION Our findings show that periodic screening for pHPT in patients with PTs may be useful to detect MEN1 syndrome; thus, it is recommended in those patients genetic analysis of MEN1 gene and an additional search of related tumors. By contrast, our results suggest that CDKN1B and AIP mutations do not seem to play a relevant role in the pathogenesis of MEN1.
Collapse
Affiliation(s)
- Sabrina S P Damianse
- Service of Endocrinology, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Postgraduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luís, Brazil
| | - Gilvan C Nascimento
- Service of Endocrinology, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil
| | - Viviane C C Rocha
- Service of Endocrinology, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil
| | - Ana Gisélia P A C Nascimento
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Pathology Unit, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA), São Luís, Brazil
| | - Nadja C Gaido
- Pathology Unit, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA), São Luís, Brazil
| | - Rossana S S Azulay
- Service of Endocrinology, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil
| | - Wellyandra C Dos Santos
- Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Postgraduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luís, Brazil
| | - Vandilson P Rodrigues
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Department of Morphology, Federal University of Maranhão (UFMA), São Luís, Brazil
| | - Ana Rosa P Quidute
- Department of Physiology and Pharmacology, Federal University of Ceará (UFC), Fortaleza, Brazil
| | - Marcelo Magalhães
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research and Extension Nucleus (NUPE), UNDB University Center, São Luís, Brazil
| | - Manuel Dos S Faria
- Service of Endocrinology, Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil; Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), São Luís, Brazil; Clinical Research Center (CEPEC), Presidente Dutra University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luís, Brazil.
| |
Collapse
|
37
|
Multiple Endocrine Neoplasia in Childhood: An Update on Diagnosis, Screening, Management and Treatment. ENDOCRINES 2022. [DOI: 10.3390/endocrines3010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Multiple endocrine neoplasia (MEN) is a group of heterogenous syndromes characterized by the occurrence of two or more endocrine gland tumors in a patient or related individuals in the same family. They are inherited in an autosomal dominant fashion and are highly penetrant. There are three types of MEN syndromes: MEN type 1 (MEN1), MEN type 2 (MEN2), and MEN type 4 (MEN4). MEN2 is further divided into MEN2A, MEN2B (formerly known MEN3), and familial medullary thyroid carcinoma (FMTC). Although MEN syndromes are rare, it is crucial to identify individuals at risk for potentially life-threatening neoplasias. This review article provides an update on each MEN syndrome, its genetics, diagnosis, and management in children.
Collapse
|
38
|
De Sousa SMC, Carroll RW, Henderson A, Burgess J, Clifton-Bligh RJ. A contemporary clinical approach to genetic testing for heritable hyperparathyroidism syndromes. Endocrine 2022; 75:23-32. [PMID: 34773560 DOI: 10.1007/s12020-021-02927-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 10/23/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE The improved access and affordability of next generation sequencing has facilitated the clinical use of gene panel testing to test concurrently patients for multiple heritable hyperparathyroidism syndromes. However, there is little guidance as to which patients should be selected for gene panel testing and which genes should be included in such panels. In this review, we provide a practical approach to considering, interpreting and managing genetic testing for familial primary hyperparathyroidism (PHPT) syndromes and familial hypocalciuric hypercalcaemia (FHH) in patients with PTH-dependent hypercalcaemia. We discuss known genes implicated in PHPT and FHH, testing criteria and yields, pre-test counselling, laboratory considerations, and post-test management. METHODS In addition to reviewing the literature, we conducted audits of local genetic testing data to examine the real-world yield of genetic testing in patients with PTH-dependent hypercalcaemia. RESULTS Our local audits revealed a positive genetic testing rate of 15-26% in patients with suspected hyperparathyroidism syndromes. CONCLUSION Based on the particular testing criteria met, affected patients should be tested for variants in the genes currently implicated in PHPT (MEN1, CDC73, RET, CDKN1B, GCM2, CASR) and/or FHH (CASR, GNA11, AP2S1). Patients should be provided with pre- and post-test counselling, including consideration of potential implications for family members.
Collapse
Affiliation(s)
- Sunita M C De Sousa
- Endocrine & Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia.
- South Australian Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
| | - Richard W Carroll
- Endocrine, Diabetes, and Research Centre, Wellington Regional Hospital, Wellington, New Zealand
| | - Alex Henderson
- Wellington Hospital, Genetic Health Service New Zealand, Wellington, New Zealand
| | - John Burgess
- Department of Diabetes and Endocrinology, Royal Hobart Hospital, Hobart, TAS, Australia
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Roderick J Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
39
|
Lavezzi E, Brunetti A, Smiroldo V, Nappo G, Pedicini V, Vitali E, Trivellin G, Mazziotti G, Lania A. Case Report: New CDKN1B Mutation in Multiple Endocrine Neoplasia Type 4 and Brief Literature Review on Clinical Management. Front Endocrinol (Lausanne) 2022; 13:773143. [PMID: 35355569 PMCID: PMC8959648 DOI: 10.3389/fendo.2022.773143] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/25/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The fourth type of multiple endocrine neoplasia (MEN) is known as a rare variant of MEN presenting a MEN1-like phenotype and originating from a germline mutation in CDKN1B. However, due to the small number of cases documented in the literature, the peculiar clinical features of MEN4 are still largely unknown, and clear indications about the clinical management of these patients are currently lacking. In order to widen our knowledge on MEN4 and to better typify the clinical features of this syndrome, we present two more cases of subjects with MEN4, and through a review of the current literature, we provide some possible indications on these patients' management. CASE PRESENTATION The first report is about a man who was diagnosed with a metastatic ileal G2-NET at the age of 34. Genetic analysis revealed the mutation p.I119T (c.356T>C) of exon 1 of CDKN1B, a mutation already reported in the literature in association with early-onset pituitary adenomas. The second report is about a 76-year-old woman with a multifocal pancreatic G1-NET. Genetic analysis identified the CDKN1B mutation c.482C>G (p.S161C), described here for the first time in association with MEN4 and currently classified as a variant of uncertain significance. Both patients underwent biochemical and imaging screening for MEN1-related diseases without any pathological findings. CONCLUSIONS According to the cases reported in the literature, hyperparathyroidism is the most common clinical feature of MEN4, followed by pituitary adenoma and neuroendocrine tumors. However, MEN4 appears to be a variant of MEN with milder clinical features and later onset. Therefore, these patients might need a different and personalized approach in clinical management and a peculiar screening and follow-up strategy.
Collapse
Affiliation(s)
- Elisabetta Lavezzi
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
- *Correspondence: Elisabetta Lavezzi,
| | - Alessandro Brunetti
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Valeria Smiroldo
- Oncology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Gennaro Nappo
- Pancreatic Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | | | - Eleonora Vitali
- Endocrinology Unit and Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giampaolo Trivellin
- Endocrinology Unit and Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Gherardo Mazziotti
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Andrea Lania
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
40
|
Spada A, Mantovani G, Lania AG, Treppiedi D, Mangili F, Catalano R, Carosi G, Sala E, Peverelli E. Pituitary Tumors: Genetic and Molecular Factors Underlying Pathogenesis and Clinical Behavior. Neuroendocrinology 2022; 112:15-33. [PMID: 33524974 DOI: 10.1159/000514862] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022]
Abstract
Pituitary neuroendocrine tumors (PitNETs) are the most common intracranial neoplasms. Although generally benign, they can show a clinically aggressive course, with local invasion, recurrences, and resistance to medical treatment. No universally accepted biomarkers of aggressiveness are available yet, and predicting clinical behavior of PitNETs remains a challenge. In rare cases, the presence of germline mutations in specific genes predisposes to PitNET formation, as part of syndromic diseases or familial isolated pituitary adenomas, and associates to more aggressive, invasive, and drug-resistant tumors. The vast majority of cases is represented by sporadic PitNETs. Somatic mutations in the α subunit of the stimulatory G protein gene (gsp) and in the ubiquitin-specific protease 8 (USP8) gene have been recognized as pathogenetic factors in sporadic GH- and ACTH-secreting PitNETs, respectively, without an association with a worse clinical phenotype. Other molecular factors have been found to significantly affect PitNET drug responsiveness and invasive behavior. These molecules are cytoskeleton and/or scaffold proteins whose alterations prevent proper functioning of the somatostatin and dopamine receptors, targets of medical therapy, or promote the ability of tumor cells to invade surrounding tissues. The aim of the present review is to provide an overview of the genetic and molecular alterations that can contribute to determine PitNET clinical behavior. Understanding subcellular mechanisms underlying pituitary tumorigenesis and PitNET clinical phenotype will hopefully lead to identification of new potential therapeutic targets and new markers predicting the behavior and the response to therapeutic treatments of PitNETs.
Collapse
Affiliation(s)
- Anna Spada
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea G Lania
- Endocrinology, Diabetology and Medical Andrology Unit, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Donatella Treppiedi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Mangili
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Rosa Catalano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giulia Carosi
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Sala
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy,
| |
Collapse
|
41
|
Abstract
Primary hyperparathyroidism (PHPT) is a commonly encountered clinical problem and occurs as part of an inherited disorder in ∼10% of patients. Several features may alert the clinician to the possibility of a hereditary PHPT disorder (eg, young age of disease onset) whilst establishing any relevant family history is essential to the clinical evaluation and will help inform the diagnosis. Genetic testing should be offered to patients at risk of a hereditary PHPT disorder, as this may improve management and allow the identification and investigation of other family members who may also be at risk of disease.
Collapse
Affiliation(s)
- Paul J Newey
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Jacqui Wood Cancer Centre, James Arrott Drive, Dundee, Scotland DD1 9SY, UK.
| |
Collapse
|
42
|
Medina JE, Randolph GW, Angelos P, Zafereo ME, Tufano RP, Kowalski LP, Montenegro FLM, Owen RP, Khafif A, Suárez C, Shaha AR, Rodrigo JP, Krempl GA, Rinaldo A, Silver CE, Ferlito A. Primary hyperparathyroidism: Disease of diverse genetic, symptomatic, and biochemical phenotypes. Head Neck 2021; 43:3996-4009. [PMID: 34541734 DOI: 10.1002/hed.26861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic, symptomatic, and biochemical heterogeneity of patients with primary hyperparathyroidism (PHPT) has become apparent in recent years. An in-depth, evidence-based review of the phenotypes of PHPT was conducted. This review was intended to provide the resulting information to surgeons who operate on patients with hyperparathyroidism. This review revealed that the once relatively clear distinction between familial and sporadic PHPT has become more challenging by the finding of various germline mutations in patients with seemingly sporadic PHPT. On the one hand, the genetic and clinical characteristics of some syndromes in which PHPT is an important component are now better understood. On the other hand, knowledge is emerging about novel syndromes, such as the rare multiple endocrine neoplasia type IV (MEN4), in which PHPT occurs frequently. It also revealed that, currently, the classical array of symptoms of PHPT is seen rarely upon initial presentation for evaluation. More common are nonspecific, nonclassical symptoms and signs of PHPT. In areas of the world where serum calcium levels are checked routinely, most patients today are "asymptomatic" and they are diagnosed after an incidental finding of hypercalcemia; however, some of them have subclinical involvement of bones and kidneys, which is demonstrated on radiographs, ultrasound, and modern imaging techniques. Last, the review points out that there are three distinct biochemical phenotypes of PHPT. The classical phenotype in which calcium and parathyroid hormone levels are both elevated, and other disease presentations in which the serum levels of calcium or intact parathyroid hormone are normal. Today several, distinct phenotypes of the disease can be identified, and they have implications in the diagnostic evaluation and treatment of patients, as well as possible screening of relatives.
Collapse
Affiliation(s)
- Jesus E Medina
- Department of Otolaryngology and Head and Neck Surgery, The University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, USA
| | - Gregory W Randolph
- Division of Thyroid and Parathyroid Endocrine Surgery, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Peter Angelos
- Department of Surgery and MacLean Center for Clinical Medical Ethics, The University of Chicago, Chicago, Illinois, USA
| | - Mark E Zafereo
- Head and Neck Endocrine Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Ralph P Tufano
- Division of Head and Neck Endocrine Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins, Baltimore, Maryland, USA
| | - Luiz P Kowalski
- Department of Head and Neck Surgery, University of Sao Paulo Medical School, São Paulo, Brazil.,Head and Neck Surgery and Otorhinolaryngology Department, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Fabio L M Montenegro
- Department of Head and Neck Surgery, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Randall P Owen
- Division of Surgical Oncology, Department of Surgery, Mount Sinai School of Medicine, New York, New York, USA
| | - Avi Khafif
- Head and Neck Surgery and Oncology Unit, A.R.M. Center for Advanced Otolaryngology Head and Neck Surgery, Assuta Medical Center, Tel Aviv, Israel
| | - Carlos Suárez
- Instituto de Investigación Sanitaria del Principado de Asturias, IUOPA, CIBERONC, Oviedo, Spain
| | - Ashok R Shaha
- Head and Neck Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Juan P Rodrigo
- University of Oviedo, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Greg A Krempl
- Department of Otolaryngology and Head and Neck Surgery, The University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, USA
| | | | - Carl E Silver
- Department of Surgery, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Alfio Ferlito
- International Head and Neck Scientific Group, Padua, Italy
| |
Collapse
|
43
|
Li Y, Tan YQ, Tang ZX, Liao QH, Guo ZQ, Lai KB, Wang R, Chen YH. Multiple endocrine neoplasia 2A with RET mutation p.Cys611Tyr: A case report. Medicine (Baltimore) 2021; 100:e26230. [PMID: 34087905 PMCID: PMC8183697 DOI: 10.1097/md.0000000000026230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Multiple endocrine neoplasia 2A (MEN2A) is a rare autosomal-dominant genetic syndrome, frequently misdiagnosed or neglected clinically, resulting in delayed therapy to patients. PATIENT CONCERNS A 47-year-old Chinese male patient underwent laparoscopic right adrenal tumorectomy, and postoperative pathology confirmed the tumor as pheochromocytoma (PHEO). He was readmitted to the department of endocrinology and metabolism due to constant increase in carcinoembryonic antigen (CEA) at 5 months after the operation. DIAGNOSIS The patient was confirmed with medullary thyroid carcinoma (MTC), multiple neck lymph node metastasis, and pituitary microadenoma. The p.Cys611Tyr (c.1832G>A, C611Y) mutation was detected. Therefore, he was diagnosed with MEN2A. INTERVENTIONS He underwent total thyroidectomy. The gene-sequencing analysis of his family was conducted, and the C611Y mutation was detected in his daughter. OUTCOMES The level of carcinoembryonic antigen decreased significantly after thyroidectomy in this patient. Long-term follow-up management was conducted. Elevated serum calcitonin and bilateral thyroid nodules were found in his 13-year-old daughter. Thus, MEN2A was highly suspected and she was suggested to undergo total thyroidectomy. CONCLUSION Patients with MEN2A should be screened regularly and managed by a multidisciplinary team.
Collapse
|
44
|
Hu X, Guan J, Wang Y, Shi S, Song C, Li ZP, Feng ST, Chen J, Luo Y. A narrative review of multiple endocrine neoplasia syndromes: genetics, clinical features, imaging findings, and diagnosis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:944. [PMID: 34350259 PMCID: PMC8263874 DOI: 10.21037/atm-21-1165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022]
Abstract
Objective We aimed to provide ideas for clinicians, especially radiologists, for the diagnosis of multiple endocrine neoplasia (MEN) syndromes. Background MEN syndromes include MEN1, MEN2, and MEN4 and usually involve 2 or more endocrine tumors. The MEN syndromes are a group of euchromatic dominant genetic diseases, and the main genes involved include MEN1 (MEN1), RET (MEN2), and CDKN1B (MEN4). Methods In this article, involving 8 cases (4 cases of MEN1, 2 cases of MEN2A, 1 case of MEN2B, 1 case of MEN4) from our center, we introduced the disease spectrum, clinical manifestations (especially imaging findings), and related genes involved in each type of MEN syndromes. We also discussed the differential diagnosis between MEN and sporadic tumors and emphasized that MEN should be screened and the relevant required examinations. Conclusions Considering that MEN syndromes involve multiple endocrine gland tumors and nonendocrine organ diseases, it is very important to identify potential patients early and perform multiple examinations on them, including biochemical and multitype, and multisite imaging examinations according to the disease spectrum of each type. Considering that this is a group of genetic diseases, both interviewing patients about their family history and genetic testing are also very important. Only in this way can a comprehensive and accurate diagnosis be made, enabling patients to receive appropriate treatment and improve their prognosis.
Collapse
Affiliation(s)
- Xuefang Hu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian Guan
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yangdi Wang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Siya Shi
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chenyu Song
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zi-Ping Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jie Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yanji Luo
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Al-Salameh A, Cadiot G, Calender A, Goudet P, Chanson P. Clinical aspects of multiple endocrine neoplasia type 1. Nat Rev Endocrinol 2021; 17:207-224. [PMID: 33564173 DOI: 10.1038/s41574-021-00468-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 01/31/2023]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a rare syndrome characterized by the co-occurrence of primary hyperparathyroidism, duodenopancreatic neuroendocrine tumours (NETs) and/or pituitary adenomas. MEN1 can predispose patients to other endocrine and non-endocrine tumours, such as cutaneous tumours, central nervous system tumours and breast cancer. Endocrine tumours in patients with MEN1 differ from sporadic tumours in that they have a younger age at onset, present as multiple tumours in the same organ and have a different clinical course. Therefore, patients with overt MEN1 and those who carry a MEN1 mutation should be offered tailored biochemical and imaging screening to detect tumours and evaluate their progression over time. Fortunately, over the past 10 years, knowledge about the clinical phenotype of these tumours has markedly progressed, thanks to the implementation of national registries, particularly in France and the Netherlands. This Review provides an update on the clinical management of MEN1-related tumours. Epidemiology, the clinical picture, diagnostic work-up and the main lines of treatment for MEN1-related tumours are summarized. Controversial therapeutic aspects and issues that still need to be addressed are also discussed. Moreover, special attention is given to MEN1 manifestations in children and adolescents.
Collapse
Affiliation(s)
- Abdallah Al-Salameh
- Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l'Hypophyse, Le Kremlin-Bicêtre, France
- Service d'Endocrinologie, Maladies Métaboliques et Nutrition, CHU Amiens Picardie, Amiens, France
| | - Guillaume Cadiot
- Service d'Hépato-Gastro-Entérologie et de Cancérologie Digestive, Hôpital Robert Debré, Reims, France
| | - Alain Calender
- Unité Médicale des Cancers et Maladies Multifactorielles, Service de Génétique, Hospices Civils de Lyon, Lyon, France
| | - Pierre Goudet
- Service de Chirurgie Endocrinienne, Hôpital du Bocage, Dijon, France
| | - Philippe Chanson
- Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l'Hypophyse, Le Kremlin-Bicêtre, France.
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre, France.
| |
Collapse
|
46
|
Genetics of Acromegaly and Gigantism. J Clin Med 2021; 10:jcm10071377. [PMID: 33805450 PMCID: PMC8036715 DOI: 10.3390/jcm10071377] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
Growth hormone (GH)-secreting pituitary tumours represent the most genetically determined pituitary tumour type. This is true both for germline and somatic mutations. Germline mutations occur in several known genes (AIP, PRKAR1A, GPR101, GNAS, MEN1, CDKN1B, SDHx, MAX) as well as familial cases with currently unknown genes, while somatic mutations in GNAS are present in up to 40% of tumours. If the disease starts before the fusion of the epiphysis, then accelerated growth and increased final height, or gigantism, can develop, where a genetic background can be identified in half of the cases. Hereditary GH-secreting pituitary adenoma (PA) can manifest as isolated tumours, familial isolated pituitary adenoma (FIPA) including cases with AIP mutations or GPR101 duplications (X-linked acrogigantism, XLAG) or can be a part of systemic diseases like multiple endocrine neoplasia type 1 or type 4, McCune-Albright syndrome, Carney complex or phaeochromocytoma/paraganglioma-pituitary adenoma association. Family history and a search for associated syndromic manifestations can help to draw attention to genetic causes; many of these are now tested as part of gene panels. Identifying genetic mutations allows appropriate screening of associated comorbidities as well as finding affected family members before the clinical manifestation of the disease. This review focuses on germline and somatic mutations predisposing to acromegaly and gigantism.
Collapse
|
47
|
Brandi ML, Agarwal SK, Perrier ND, Lines KE, Valk GD, Thakker RV. Multiple Endocrine Neoplasia Type 1: Latest Insights. Endocr Rev 2021; 42:133-170. [PMID: 33249439 PMCID: PMC7958143 DOI: 10.1210/endrev/bnaa031] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Indexed: 02/06/2023]
Abstract
Multiple endocrine neoplasia type 1 (MEN1), a rare tumor syndrome that is inherited in an autosomal dominant pattern, is continuing to raise great interest for endocrinology, gastroenterology, surgery, radiology, genetics, and molecular biology specialists. There have been 2 major clinical practice guidance papers published in the past 2 decades, with the most recent published 8 years ago. Since then, several new insights on the basic biology and clinical features of MEN1 have appeared in the literature, and those data are discussed in this review. The genetic and molecular interactions of the MEN1-encoded protein menin with transcription factors and chromatin-modifying proteins in cell signaling pathways mediated by transforming growth factor β/bone morphogenetic protein, a few nuclear receptors, Wnt/β-catenin, and Hedgehog, and preclinical studies in mouse models have facilitated the understanding of the pathogenesis of MEN1-associated tumors and potential pharmacological interventions. The advancements in genetic diagnosis have offered a chance to recognize MEN1-related conditions in germline MEN1 mutation-negative patients. There is rapidly accumulating knowledge about clinical presentation in children, adolescents, and pregnancy that is translatable into the management of these very fragile patients. The discoveries about the genetic and molecular signatures of sporadic neuroendocrine tumors support the development of clinical trials with novel targeted therapies, along with advancements in diagnostic tools and surgical approaches. Finally, quality of life studies in patients affected by MEN1 and related conditions represent an effort necessary to develop a pharmacoeconomic interpretation of the problem. Because advances are being made both broadly and in focused areas, this timely review presents and discusses those studies collectively.
Collapse
Affiliation(s)
| | | | - Nancy D Perrier
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Gerlof D Valk
- University Medical Center Utrecht, CX Utrecht, the Netherlands
| | | |
Collapse
|
48
|
Volante M, Mete O, Pelosi G, Roden AC, Speel EJM, Uccella S. Molecular Pathology of Well-Differentiated Pulmonary and Thymic Neuroendocrine Tumors: What Do Pathologists Need to Know? Endocr Pathol 2021; 32:154-168. [PMID: 33641055 PMCID: PMC7960615 DOI: 10.1007/s12022-021-09668-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
Thoracic (pulmonary and thymic) neuroendocrine tumors are well-differentiated epithelial neuroendocrine neoplasms that are classified into typical and atypical carcinoid tumors based on mitotic index cut offs and presence or absence of necrosis. This classification scheme is of great prognostic value but designed for surgical specimens, only. Deep molecular characterization of thoracic neuroendocrine tumors highlighted their difference with neuroendocrine carcinomas. Neuroendocrine tumors of the lung are characterized by a low mutational burden, and a high prevalence of mutations in chromatin remodeling and histone modification-related genes, whereas mutations in genes frequently altered in neuroendocrine carcinomas are rare. Molecular profiling divided thymic neuroendocrine tumors into three clusters with distinct clinical outcomes and characterized by a different average of copy number instability. Moreover, integrated histopathological, molecular and clinical evidence supports the existence of a grey zone category between neuroendocrine tumors (carcinoid tumors) and neuroendocrine carcinomas. Indeed, cases with well differentiated morphology but mitotic/Ki-67 indexes close to neuroendocrine carcinomas have been increasingly recognized. These are characterized by specific molecular profiles and have an aggressive clinical behavior. Finally, thoracic neuroendocrine tumors may arise in the background of genetic susceptibility, being MEN1 syndrome the well-defined familial form. However, pathologists should be aware of rarer germline variants that are associated with the concurrence of neuroendocrine tumors of the lung or their precursors (such as DIPNECH) with other neoplasms, including but not limited to breast carcinomas. Therefore, genetic counseling for all young patients with thoracic neuroendocrine neoplasia and/or any patient with pathological evidence of neuroendocrine cell hyperplasia-to-neoplasia progression sequence or multifocal disease should be considered.
Collapse
Affiliation(s)
- Marco Volante
- Department of Oncology, University of Turin, Turin, Italy.
| | - Ozgur Mete
- Departments of Pathology, University Healthy Network and University of Toronto, Toronto, Canada
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Ernst Jan M Speel
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Silvia Uccella
- Dept. of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
49
|
Brock P, Bustamante Alvarez J, Mortazavi A, Roychowdhury S, Phay J, Khawaja RA, Shah MH, Konda B. Co-occurrence of multiple endocrine neoplasia type 4 and spinal neurofibromatosis: a case report. Fam Cancer 2021; 19:189-192. [PMID: 32052251 DOI: 10.1007/s10689-019-00152-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Multiple Endocrine Neoplasia (MEN) type 4 is a rare genetic condition that results from variants of the CDKN1B gene and predisposes individuals to develop endocrine tumors. Spinal neurofibromatosis (SNF) is an uncommon subtype of neurofibromatosis type 1 (NF1) characterized by bilateral neurofibromas of all spinal roots. Here we report a case of the co-occurrence of these syndromes, which has not yet been described in the literature. A male in his 60s presented with Gleason 5 + 4 localized prostate adenocarcinoma treated with radical prostatectomy. Two years later, he developed liver and bone metastasis consistent with trans-differentiation into small cell carcinoma. He developed hypercalcemia due to primary hyperparathyroidism from a parathyroid adenoma treated surgically. His family history was significant for a first-degree relative with a clinical diagnosis of NF1 and several second-degree relatives with multiple café-au-lait macules. Spine MRI showed multiple bilateral neurofibromas. Germline genetic testing showed a pathogenic variant in the CDKN1B gene, a variant in the NF1 gene, and a normal MEN1 gene. In this rare case of MEN4 and SNF, the patient was asymptomatic for much of his life. In addition to parathyroid adenoma and spinal neurofibromas, he had prostate adenocarcinoma with trans-differentiation into metastatic small cell cancer. Whether this diagnosis was coincidental or related to an emerging phenotype remains to be elucidated.
Collapse
Affiliation(s)
- Pamela Brock
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, 2012 Kenny Rd, Columbus, OH, 43221, USA
| | - Jean Bustamante Alvarez
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, A440 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, A440 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - Sameek Roychowdhury
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, A440 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - John Phay
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, 410 W 10th Ave, Columbus, OH, 43210, USA
| | - Raheela A Khawaja
- Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University, 1581 Dodd Dr, Columbus, OH, 43210, USA
| | - Manisha H Shah
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, A440 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA
| | - Bhavana Konda
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, A440 Starling-Loving Hall, 320 W 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
50
|
Lim JY, Pommier RF. Clinical Features, Management, and Molecular Characteristics of Familial Small Bowel Neuroendocrine Tumors. Front Endocrinol (Lausanne) 2021; 12:622693. [PMID: 33732215 PMCID: PMC7959745 DOI: 10.3389/fendo.2021.622693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Small bowel neuroendocrine tumors are rare tumors with an increasing incidence over the last several decades. Early detection remains challenging because patients commonly develop symptoms late in the disease course, often after the tumors have metastasized. Although these tumors were thought to arise from sporadic genetic mutations, large epidemiological studies strongly support genetic predisposition and increased risk of disease in affected families. Recent studies of familial small bowel neuroendocrine tumors have identified several novel genetic mutations. Screening for familial small bowel neuroendocrine tumors can lead to earlier diagnosis and improved patient outcomes. This review aims to summarize the current knowledge of molecular changes seen in familial small bowel neuroendocrine tumors, identify clinical features specific to familial disease, and provide strategies for screening and treatment.
Collapse
|