1
|
Sarkadi B, Liko I, Nyiro G, Igaz P, Butz H, Patocs A. Analytical Performance of NGS-Based Molecular Genetic Tests Used in the Diagnostic Workflow of Pheochromocytoma/Paraganglioma. Cancers (Basel) 2021; 13:4219. [PMID: 34439371 PMCID: PMC8392134 DOI: 10.3390/cancers13164219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/30/2022] Open
Abstract
Next Generation Sequencing (NGS)-based methods are high-throughput and cost-effective molecular genetic diagnostic tools. Targeted gene panel and whole exome sequencing (WES) are applied in clinical practice for assessing mutations of pheochromocytoma/paraganglioma (PPGL) associated genes, but the best strategy is debated. Germline mutations of at the least 18 PPGL genes are present in approximately 20-40% of patients, thus molecular genetic testing is recommended in all cases. We aimed to evaluate the analytical and clinical performances of NGS methods for mutation detection of PPGL-associated genes. WES (three different library preparation and bioinformatics workflows) and an in-house, hybridization based gene panel (endocrine-onco-gene-panel- ENDOGENE) was evaluated on 37 (20 WES and 17 ENDOGENE) samples with known variants. After optimization of the bioinformatic workflow, 61 additional samples were tested prospectively. All clinically relevant variants were validated with Sanger sequencing. Target capture of PPGL genes differed markedly between WES platforms and genes tested. All known variants were correctly identified by all methods, but methods of library preparations, sequencing platforms and bioinformatical settings significantly affected the diagnostic accuracy. The ENDOGENE panel identified several pathogenic mutations and unusual genotype-phenotype associations suggesting that the whole panel should be used for identification of genetic susceptibility of PPGL.
Collapse
Affiliation(s)
- Balazs Sarkadi
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
| | - Istvan Liko
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Bionics Innovation Center, H-1089 Budapest, Hungary;
| | - Gabor Nyiro
- Bionics Innovation Center, H-1089 Budapest, Hungary;
- MTA-SE Molecular Medicine Research Group, Eotvos Lorand Research Network, H-1083 Budapest, Hungary;
| | - Peter Igaz
- MTA-SE Molecular Medicine Research Group, Eotvos Lorand Research Network, H-1083 Budapest, Hungary;
- Department of Endocrinology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Henriett Butz
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Department of Laboratory Medicine, Semmelweis University, H-1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, H-1122 Budapest, Hungary
| | - Attila Patocs
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Bionics Innovation Center, H-1089 Budapest, Hungary;
- Department of Laboratory Medicine, Semmelweis University, H-1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, H-1122 Budapest, Hungary
| |
Collapse
|
2
|
Shi Z, Kong X, Li C, Liu H, Aliagan AI, Liu L, Shi Y, Shi X, Ma B, Jin R, Wang S, Pan D, Tang J. Bioinformatic analysis of differentially expressed genes as prognostic markers in pheochromocytoma and paraganglioma tumors. Genes Genet Syst 2021; 96:55-69. [PMID: 34039789 DOI: 10.1266/ggs.20-00057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The pathogenesis of pheochromocytoma and paraganglioma (PCPG) catecholamine-producing tumors is exceedingly complicated. Here, we sought to identify important genes affecting the prognosis and survival rate of patients suffering from PCPG. We analyzed 95 samples obtained from two microarray data series, GSE19422 and GSE60459, from the Gene Expression Omnibus (GEO) repository. First, differentially expressed genes (DEGs) were identified by comparing 87 PCPG tumor samples and eight normal adrenal tissue samples using R language. The GEO2R tool and Venn diagram software were applied to the Database for Annotation, Visualization and Integrated Discovery (DAVID) to analyze Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO). We further employed Cytoscape with the Molecular Complex Detection (MCODE) tool to make protein-protein interactions visible for the Search Tool for Retrieval of Interacting Genes (STRING). These procedures resulted in 30 candidate DEGs, which were subjected to Kaplan-Meier analysis and validated by Gene Expression Profiling Interactive Analysis (GEPIA) to determine their influence on overall survival rate. Finally, we identified ALDH3A2 and AKR1B1, two genes in the glycerolipid metabolism pathway, as being particularly enriched in PCPG tumors and correlated with T and B tumor-infiltrating immune cells. Our results suggest that these two DEGs are closely associated with the prognosis of malignant PCPG tumors.
Collapse
Affiliation(s)
- Zhen Shi
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University.,Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Xiaodi Kong
- Department of Urology, Xiangya Hospital, Central South University
| | - Cheng Li
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Hui Liu
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Abdulhafiz Imam Aliagan
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Li Liu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Yue Shi
- School of Mechanical Engineering, Northwestern Polytechnical University
| | - Xiao Shi
- The Third Xiangya Hospital, Central South University
| | - Binbin Ma
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Ruiqi Jin
- The Third Xiangya Hospital, Central South University
| | - Shizhuo Wang
- College of Life Science and Technology, Beijing University of Chemical Technology
| | - Ding Pan
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Juyu Tang
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| |
Collapse
|
3
|
Butz H, Blair J, Patócs A. Molecular genetic testing strategies used in diagnostic flow for hereditary endocrine tumour syndromes. Endocrine 2021; 71:641-652. [PMID: 33570725 PMCID: PMC8016766 DOI: 10.1007/s12020-021-02636-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/18/2021] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Although current guidelines prefer the use of targeted testing or small-scale gene panels for identification of genetic susceptibility of hereditary endocrine tumour syndromes, next generation sequencing based strategies have been widely introduced into every day clinical practice. The application of next generation sequencing allows rapid testing of multiple genes in a cost effective manner. Increasing knowledge about these techniques and the demand from health care providers and society, shift the molecular genetic testing towards using high-throughput approaches. PURPOSE In this expert opinion, the authors consider the molecular diagnostic workflow step by step, evaluating options and challenges of gathering family information, pre- and post-test genetic counselling, technical and bioinformatical analysis related issues and difficulties in clinical interpretation focusing on molecular genetic testing of hereditary endocrine tumour syndromes. RESULT AND CONCLUSION Considering all these factors, a diagnostic genetic workflow is also proposed for selection of the best approach for testing of patients with hereditary genetic tumour syndromes in order to minimalize difficult interpretation, unwanted patient anxiety, unnecessary medical interventions and cost. There are potential benefits of utilizing high throughput approaches however, important limitations have to be considered and should discussed towards the clinicians and patients.
Collapse
Affiliation(s)
- Henriett Butz
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
- Hereditary Cancers Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Jo Blair
- Alder Hey Children's Hospital-NHS Foundation Trust, Liverpool, United Kingdom
| | - Attila Patócs
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary.
- Hereditary Cancers Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary.
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary.
- Semmelweis University, Budapest, Hungary.
| |
Collapse
|
4
|
de Tersant M, Généré L, Freyçon C, Villebasse S, Abbas R, Barlier A, Bodet D, Corradini N, Defachelles AS, Entz-Werle N, Fouquet C, Galmiche L, Gandemer V, Lacour B, Mansuy L, Orbach D, Pluchart C, Réguerre Y, Rigaud C, Sarnacki S, Sirvent N, Stephan JL, Thebaud E, Gimenez-Roqueplo AP, Brugières L. Pheochromocytoma and Paraganglioma in Children and Adolescents: Experience of the French Society of Pediatric Oncology (SFCE). J Endocr Soc 2020; 4:bvaa039. [PMID: 32432211 PMCID: PMC7217277 DOI: 10.1210/jendso/bvaa039] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 04/01/2020] [Indexed: 11/19/2022] Open
Abstract
Purpose The purpose of this work is to assess the clinical outcome of pediatric patients diagnosed with pheochromocytoma and paraganglioma (PPGL) detected in France since 2000. Methods A retrospective multicenter study was conducted that included all patients younger than 18 years with PPGL diagnosed in France between 2000 and 2016. Patients were identified from 4 different sources: the National Registry of Childhood Solid Tumors, the French Pediatric Rare Tumors Database, the French registry of succinate dehydrogenase (SDH)-related hereditary paraganglioma, and the nationwide TenGen network. Results Among 113 eligible patients, 81 children with available data were enrolled (41 with adrenal and 40 with extra-adrenal PPGL). At diagnosis, 11 had synchronous metastases. After a median follow-up of 53 months, 27 patients experienced a new event (n = 7 second PPGL, n = 1 second paraganglioma [PGL], n = 8 local recurrences, n = 10 metastatic relapses, n = 1 new tumor) and 2 patients died of their disease. The 3- and 10-year event-free survival rates were 80% (71%-90%) and 39% (20%-57%),respectively, whereas the overall survival rate was 97% (93%-100%)at 3 and 10 years. A germline mutation in one PPGL-susceptibility gene was identified in 53 of the 68 (77%) patients who underwent genetic testing (SDHB [n = 25], VHL [n = 21], RET [n = 2], HIF2A [n = 2], SDHC [n = 1], SDHD [n = 1], NF1 [n = 1]). Incomplete resection and synchronous metastases were associated with higher risk of events (P = .011, P = .004), but presence of a germline mutation was not (P = .11). Conclusions Most pediatric PPGLs are associated with germline mutations and require specific follow-up because of the high risk of tumor recurrence.
Collapse
Affiliation(s)
- Marie de Tersant
- Centre de Cancérologie Gustave Roussy, Département de Cancérologie de l'Enfant et de l'Adolescent, Paris-Saclay University Villejuif, France
| | - Lucile Généré
- Institut d'Hématologie et d'oncologie pédiatrique, Lyon, France
| | - Claire Freyçon
- Centre Hospitalier Universitaire Grenoble Alpes, Immuno-hémato-oncologie pédiatrique, La Tronche, France
| | - Sophie Villebasse
- Centre de Cancérologie Gustave Roussy, Département de Cancérologie de l'Enfant et de l'Adolescent, Paris-Saclay University Villejuif, France
| | - Rachid Abbas
- Centre de Cancérologie Gustave Roussy, Service de Biostatistique et d'Epidémiologie, Paris-Saclay University Villejuif, France
| | - Anne Barlier
- Aix Marseille Université, APHM, INSERM, MMG, Laboratory of Molecular Biology Hospital La Conception, Marseille Cedex 5, France
| | - Damien Bodet
- CHU de Caen, Hémato- Immuno-Oncologie Pédiatrique, Caen, France
| | | | | | - Natacha Entz-Werle
- Hôpitaux Universitaire de Strasbourg, Onco-Hématologie Pédiatrique, Strasbourg, France
| | - Cyrielle Fouquet
- Groupe Hospitalier Pellegrin, Unité d'Oncologie et Hématologie Pédiatriques, Bordeaux, France
| | - Louise Galmiche
- Assistance Publique Hôpitaux de Paris, Hôpital Necker Enfants Malades, Anatomie Pathologique, Paris, France
| | - Virginie Gandemer
- Hôpital Sud, CHU de Rennes, Hémato-Oncologie Pédiatrique, Rennes, France
| | - Brigitte Lacour
- Registre National des Tumeurs Solides de l'Enfant, CHU de Nancy, 54500 Vandoeuvre-Lès-Nancy, France; Inserm U1153, Centre de Recherche Epidémiolgie et StatistiqueS (CRESS), Université de Paris, Equipe d'Epidemiologie des cancers de l'enfant et de l'adolescent (EPICEA), Paris, France
| | - Ludovic Mansuy
- CHU de Nancy-Hôpital de Brabois, Service d'hémato-oncologie pédiatrique, Vandoeuvre-lès-Nancy Cedex, France
| | - Daniel Orbach
- SIREDO Oncology Center (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Université PSL Paris, France, Paris, France.,Comité FRACTURE des tumeurs très rares pédiatriques de la SFCE
| | | | - Yves Réguerre
- CHU de Saint Denis, Service d'Oncologie et d'hématologie pédiatrique, La Réunion, France
| | - Charlotte Rigaud
- Centre de Cancérologie Gustave Roussy, Département de Cancérologie de l'Enfant et de l'Adolescent, Paris-Saclay University Villejuif, France
| | - Sabine Sarnacki
- Assistance Publique Hôpitaux de Paris, Hôpital Necker Enfants Malades, Chirurgie Pédiatrique, Paris, France
| | - Nicolas Sirvent
- CHU de Montpellier, Onco-Hématologie Pédiatrique, Montpellier, France
| | - Jean-Louis Stephan
- CHU de Saint-Etienne, Hématologie et Oncologie Pédiatrique, Hôpital Nord, Saint-Priest-en-Jarez, France
| | - Estelle Thebaud
- CHU de Nantes, Hématologie et Oncologie Pédiatrique, Nantes, France
| | - Anne-Paule Gimenez-Roqueplo
- Université de Paris, PARCC, INSERM, Paris, France.,Service de Génétique, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Laurence Brugières
- Centre de Cancérologie Gustave Roussy, Département de Cancérologie de l'Enfant et de l'Adolescent, Paris-Saclay University Villejuif, France
| |
Collapse
|
5
|
Abstract
Pheochromocytomas and extra-adrenal paragangliomas are rare neuroendocrine neoplasms with characteristic histologic and immunohistochemical features. These tumors can arise in several anatomic locations, necessitating that their diagnostic recognition extends beyond the realm of endocrine disorders. A practical and reproducible risk stratification system for these tumors is still in development. In this rapidly evolving era of molecular medicine, it is essential for pathologists to equip themselves with a framework for understanding the classification of paragangliomas and pheochromocytomas and be informed of how they might advise their colleagues with regard to prognostication and appropriate follow-up.
Collapse
Affiliation(s)
- Julie Guilmette
- Department of Pathology, Charles-Lemoyne Hospital, Sherbrooke University Affiliated Health Care Center, 3120 Boulevard Taschereau, Greenfield Park, Quebec J4V 2H1, Canada
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114-2696, USA.
| |
Collapse
|
6
|
Tuaeva NO, Falzone L, Porozov YB, Nosyrev AE, Trukhan VM, Kovatsi L, Spandidos DA, Drakoulis N, Kalogeraki A, Mamoulakis C, Tzanakakis G, Libra M, Tsatsakis A. Translational Application of Circulating DNA in Oncology: Review of the Last Decades Achievements. Cells 2019; 8:E1251. [PMID: 31615102 PMCID: PMC6829588 DOI: 10.3390/cells8101251] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/30/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
In recent years, the introduction of new molecular techniques in experimental and clinical settings has allowed researchers and clinicians to propose circulating-tumor DNA (ctDNA) analysis and liquid biopsy as novel promising strategies for the early diagnosis of cancer and for the definition of patients' prognosis. It was widely demonstrated that through the non-invasive analysis of ctDNA, it is possible to identify and characterize the mutational status of tumors while avoiding invasive diagnostic strategies. Although a number of studies on ctDNA in patients' samples significantly contributed to the improvement of oncology practice, some investigations generated conflicting data about the diagnostic and prognostic significance of ctDNA. Hence, to highlight the relevant achievements obtained so far in this field, a clearer description of the current methodologies used, as well as the obtained results, are strongly needed. On these bases, this review discusses the most relevant studies on ctDNA analysis in cancer, as well as the future directions and applications of liquid biopsy. In particular, special attention was paid to the early diagnosis of primary cancer, to the diagnosis of tumors with an unknown primary location, and finally to the prognosis of cancer patients. Furthermore, the current limitations of ctDNA-based approaches and possible strategies to overcome these limitations are presented.
Collapse
Affiliation(s)
- Natalia O Tuaeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Luca Falzone
- Department of Biomedical and Biotechnlogical Sciences, University of Catania, 95123 Catania, Italy.
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", 80131 Naples, Italy.
| | - Yuri B Porozov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
- ITMO University, Saint Petersburg 197101, Russia.
| | - Alexander E Nosyrev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Vladimir M Trukhan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54248 Thessaloniki, Greece.
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Zografou, Greece.
| | - Alexandra Kalogeraki
- Department of Pathology-Cytopathology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, 70013 Crete, Greece.
| | - George Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Massimo Libra
- Department of Biomedical and Biotechnlogical Sciences, University of Catania, 95123 Catania, Italy.
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy.
| | - Aristides Tsatsakis
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.
| |
Collapse
|
7
|
Welander J, Łysiak M, Brauckhoff M, Brunaud L, Söderkvist P, Gimm O. Activating FGFR1 Mutations in Sporadic Pheochromocytomas. World J Surg 2018; 42:482-489. [PMID: 29159601 PMCID: PMC5762800 DOI: 10.1007/s00268-017-4320-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Introduction Pheochromocytomas are neuroendocrine tumors of the adrenal glands. Up to 40% of the cases are caused by germline mutations in one of at least 15 susceptibility genes, making them the human neoplasms with the highest degree of heritability. Recurrent somatic alterations are found in about 50% of the more common sporadic tumors with NF1 being the most common mutated gene (20–25%). In many sporadic tumors, however, a genetic explanation is still lacking. Materials and methods We investigated the genomic landscape of sporadic pheochromocytomas with whole-exome sequencing of 16 paired tumor and normal DNA samples and extended confirmation analysis in 2 additional cohorts comprising a total of 80 sporadic pheochromocytomas. Results We discovered on average 33 non-silent somatic variants per tumor. One of the recurrently mutated genes was FGFR1, encoding the fibroblast growth factor receptor 1, which was recently revealed as an oncogene in pediatric brain tumors. Including a subsequent analysis of a larger cohort, activating FGFR1 mutations were detected in three of 80 sporadic pheochromocytomas (3.8%). Gene expression microarray profiling showed that these tumors clustered with NF1-, RET,- and HRAS-mutated pheochromocytomas, indicating activation of the MAPK and PI3K-AKT signal transduction pathways. Conclusion Besides RET and HRAS, FGFR1 is only the third protooncogene found to be recurrently mutated in pheochromocytomas. The results advance our biological understanding of pheochromocytoma and suggest that somatic FGFR1 activation is an important event in a subset of sporadic pheochromocytomas. Electronic supplementary material The online version of this article (10.1007/s00268-017-4320-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jenny Welander
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Małgorzata Łysiak
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Michael Brauckhoff
- Department of Surgery, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
| | - Laurent Brunaud
- Department of Digestive, Hepato-Biliary and Endocrine Surgery, CHU Nancy - Hospital Brabois Adultes, University de Lorraine, 54511, Vandoeuvre-les-Nancy, France
| | - Peter Söderkvist
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden.
| | - Oliver Gimm
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden.,Department of Surgery, County Council of Östergötland, 58185, Linköping, Sweden
| |
Collapse
|
8
|
Neumann HP, Young WF, Krauss T, Bayley JP, Schiavi F, Opocher G, Boedeker CC, Tirosh A, Castinetti F, Ruf J, Beltsevich D, Walz M, Groeben HT, von Dobschuetz E, Gimm O, Wohllk N, Pfeifer M, Lourenço DM, Peczkowska M, Patocs A, Ngeow J, Makay Ö, Shah NS, Tischler A, Leijon H, Pennelli G, Villar Gómez de Las Heras K, Links TP, Bausch B, Eng C. 65 YEARS OF THE DOUBLE HELIX: Genetics informs precision practice in the diagnosis and management of pheochromocytoma. Endocr Relat Cancer 2018; 25:T201-T219. [PMID: 29794110 DOI: 10.1530/erc-18-0085] [Citation(s) in RCA: 40] [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: 05/14/2018] [Accepted: 05/24/2018] [Indexed: 12/21/2022]
Abstract
Although the authors of the present review have contributed to genetic discoveries in the field of pheochromocytoma research, we can legitimately ask whether these advances have led to improvements in the diagnosis and management of patients with pheochromocytoma. The answer to this question is an emphatic Yes! In the field of molecular genetics, the well-established axiom that familial (genetic) pheochromocytoma represents 10% of all cases has been overturned, with >35% of cases now attributable to germline disease-causing mutations. Furthermore, genetic pheochromocytoma can now be grouped into five different clinical presentation types in the context of the ten known susceptibility genes for pheochromocytoma-associated syndromes. We now have the tools to diagnose patients with genetic pheochromocytoma, identify germline mutation carriers and to offer gene-informed medical management including enhanced surveillance and prevention. Clinically, we now treat an entire family of tumors of the paraganglia, with the exact phenotype varying by specific gene. In terms of detection and classification, simultaneous advances in biochemical detection and imaging localization have taken place, and the histopathology of the paraganglioma tumor family has been revised by immunohistochemical-genetic classification by gene-specific antibody immunohistochemistry. Treatment options have also been substantially enriched by the application of minimally invasive and adrenal-sparing surgery. Finally and most importantly, it is now widely recognized that patients with genetic pheochromocytoma/paraganglioma syndromes should be treated in specialized centers dedicated to the diagnosis, treatment and surveillance of this rare neoplasm.
Collapse
Affiliation(s)
- Hartmut P Neumann
- Section for Preventive MedicineUniversity Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - William F Young
- Division of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, New York, USA
| | - Tobias Krauss
- Department of RadiologyMedical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jean-Pierre Bayley
- Department of Human GeneticsLeiden University Medical Center, Leiden, The Netherlands
| | - Francesca Schiavi
- Familial Cancer Clinic and OncoendocrinologyVeneto Institute of Oncology, IRCCS, Padova, Italy
| | - Giuseppe Opocher
- Familial Cancer Clinic and OncoendocrinologyVeneto Institute of Oncology, IRCCS, Padova, Italy
| | - Carsten C Boedeker
- Department of OtorhinolaryngologyHELIOS Hanseklinikum Stralsund, Stralsund, Germany
| | - Amit Tirosh
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | - Frederic Castinetti
- Department of EndocrinologyAix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Marseille, France
- Assistance Publique - Hôpitaux de Marseille (AP-HM)Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Marseille, France
| | - Juri Ruf
- Department of Nuclear MedicineFaculty of Medicine, Albert-Ludwigs-University, Freiburg, Germany
| | | | - Martin Walz
- Department of Surgery and Center of Minimally-Invasive SurgeryKliniken Essen-Mitte, Essen, Germany
| | | | - Ernst von Dobschuetz
- Section of Endocrine SurgeryClinic of General, Visceral and Thoracic Surgery, Krankenhaus Reinbek, Academic Teaching Hospital University of Hamburg, Reinbek, Germany
| | - Oliver Gimm
- Department of Clinical and Experimental MedicineFaculty of Health Sciences, Linköping University, Linköping, Sweden
- Department of SurgeryRegion Östergötland, Linköping, Sweden
| | - Nelson Wohllk
- Endocrine SectionUniversidad de Chile, Hospital del Salvador, Santiago de Chile, Chile
| | - Marija Pfeifer
- Department of EndocrinologyUniversity Medical Center Ljubljana, Ljubljana, Slovenia
| | - Delmar M Lourenço
- Endocrine Genetics UnitEndocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine (FMUSP), Endocrine Oncology Division, Institute of Cancer of the State of São Paulo, FMUSP, São Paulo, Brazil
| | | | - Attila Patocs
- HSA-SE 'Lendület' Hereditary Endocrine Tumor Research GroupHungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Joanne Ngeow
- Lee Kong Chian School of MedicineNanyang Technological University Singapore and Cancer Genetics Service, National Cancer Centre Singapore, Singapore, Singapore
| | - Özer Makay
- Division of Endocrine SurgeryDepartment of General Surgery, Ege University, Izmir, Turkey
| | - Nalini S Shah
- Department of EndocrinologySeth G S Medical College, K.E.M. Hospital, Parel, Mumbai, India
| | - Arthur Tischler
- Department of Pathology and Laboratory MedicineTufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Helena Leijon
- Department of PathologyUniversity of Helsinki, and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Gianmaria Pennelli
- Department of Medicine (DIMED)Surgical Pathology Unit, University of Padua, Padua, Italy
| | | | - Thera P Links
- Department of EndocrinologyUniversity of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Birke Bausch
- Department of Medicine IIMedical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charis Eng
- Genomic Medicine InstituteLerner Research Institute and Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Dias Pereira B, Nunes da Silva T, Bernardo AT, César R, Vara Luiz H, Pacak K, Mota-Vieira L. A Clinical Roadmap to Investigate the Genetic Basis of Pediatric Pheochromocytoma: Which Genes Should Physicians Think About? Int J Endocrinol 2018; 2018:8470642. [PMID: 29755524 PMCID: PMC5884154 DOI: 10.1155/2018/8470642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/18/2018] [Indexed: 01/06/2023] Open
Abstract
Pheochromocytoma is very rare at a pediatric age, and when it is present, the probability of a causative genetic mutation is high. Due to high costs of genetic surveys and an increasing number of genes associated with pheochromocytoma, a sequential genetic analysis driven by clinical and biochemical phenotypes is advised. The published literature regarding the genetic landscape of pediatric pheochromocytoma is scarce, which may hinder the establishment of genotype-phenotype correlations and the selection of appropriate genetic testing at this population. In the present review, we focus on the clinical phenotypes of pediatric patients with pheochromocytoma in an attempt to contribute to an optimized genetic testing in this clinical context. We describe epidemiological data on the prevalence of pheochromocytoma susceptibility genes, including new genes that are expanding the genetic etiology of this neuroendocrine tumor in pediatric patients. The clinical phenotypes associated with a higher pretest probability for hereditary pheochromocytoma are presented, focusing on differences between pediatric and adult patients. We also describe new syndromes, as well as rates of malignancy and multifocal disease associated with these syndromes and pheochromocytoma susceptibility genes published more recently. Finally, we discuss new tools for genetic screening of patients with pheochromocytoma, with an emphasis on its applicability in a pediatric population.
Collapse
Affiliation(s)
- Bernardo Dias Pereira
- Serviço de Endocrinologia e Nutrição, Hospital do Divino Espírito Santo de Ponta Delgada (EPER), Av. D. Manuel I, 9500-370 Ponta Delgada, Açores, Portugal
| | - Tiago Nunes da Silva
- Serviço de Endocrinologia e Diabetes, Hospital Garcia de Orta (EPE), Av. Torrado da Silva, 2851-951 Almada, Setúbal, Portugal
| | - Ana Teresa Bernardo
- Serviço de Cirurgia Geral, Hospital do Divino Espírito Santo de Ponta Delgada (EPER), Av. D. Manuel I, 9500-370 Ponta Delgada, Açores, Portugal
| | - Rui César
- Serviço de Endocrinologia e Nutrição, Hospital do Divino Espírito Santo de Ponta Delgada (EPER), Av. D. Manuel I, 9500-370 Ponta Delgada, Açores, Portugal
| | - Henrique Vara Luiz
- Serviço de Endocrinologia e Diabetes, Hospital Garcia de Orta (EPE), Av. Torrado da Silva, 2851-951 Almada, Setúbal, Portugal
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver NICHD, NIH, Building 10 CRC 1E-3140 10 Center Drive MSC-1109, Bethesda, MD 20892-1109, USA
| | - Luísa Mota-Vieira
- Unidade de Genética e Patologia Moleculares, Hospital do Divino Espírito Santo de Ponta Delgada (EPER), Av. D. Manuel I, 9500-370 Ponta Delgada, Açores, Portugal
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
10
|
Pillai S, Gopalan V, Lam AKY. Review of sequencing platforms and their applications in phaeochromocytoma and paragangliomas. Crit Rev Oncol Hematol 2017; 116:58-67. [DOI: 10.1016/j.critrevonc.2017.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/16/2022] Open
|
11
|
Paulo P, Pinto P, Peixoto A, Santos C, Pinto C, Rocha P, Veiga I, Soares G, Machado C, Ramos F, Teixeira MR. Validation of a Next-Generation Sequencing Pipeline for the Molecular Diagnosis of Multiple Inherited Cancer Predisposing Syndromes. J Mol Diagn 2017; 19:502-513. [PMID: 28529006 DOI: 10.1016/j.jmoldx.2017.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Despite the growing knowledge of the genetic background behind the cancers that occur in a context of hereditary predisposition, personal or family cancer history may not be clear enough to support directional gene testing. Defined targeted next-generation sequencing gene panels allow identification of the causative disease mutations of multigene syndromes and differential diagnosis for syndromes with phenotypically overlapping characteristics. Herein, we established a next-generation sequencing analysis pipeline for the molecular diagnosis of multiple inherited cancer predisposing syndromes using the commercially available target sequencing panel TruSight Cancer. To establish the analysis pipeline, we included 22 control samples with deleterious mutations covering all genes currently analyzed at our institution by standard Sanger sequencing. We tested the pipeline using 51 samples from patients with a clinical diagnosis of neurofibromatosis type 1 (NF1), 10 of which without previous molecular characterization of the causative NF1 mutations. We propose a thoroughly validated analysis pipeline that combines Isaac Enrichment, Burrows-Wheeler Aligner Enrichment, and NextGENe for the alignment and variant calling, and GeneticistAssistant for variant annotation and prioritization. This pipeline allowed the identification of disease-causing mutations in all 73 patients, including a large duplication of 37 bp in NF1. We show that high sensitivity and specificity can be achieved by using multiple bioinformatic tools for alignment and variant calling and careful variant filtering, having in mind the clinical question.
Collapse
Affiliation(s)
- Paula Paulo
- Cancer Genetics Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Pedro Pinto
- Cancer Genetics Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Ana Peixoto
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Catarina Santos
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Carla Pinto
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Patrícia Rocha
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Isabel Veiga
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Gabriela Soares
- Jacinto de Magalhães Medical Genetics Center, Centro Hospitalar do Porto, Porto, Portugal
| | - Catarina Machado
- Department of Genetics, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisboa, Portugal
| | - Fabiana Ramos
- Department of Genetics, Hospital Pediátrico Carmona da Mota, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Manuel R Teixeira
- Cancer Genetics Group, Portuguese Oncology Institute of Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal; Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal; Biomedical Sciences Institute, University of Porto, Porto, Portugal.
| |
Collapse
|
12
|
Toledo RA, Burnichon N, Cascon A, Benn DE, Bayley JP, Welander J, Tops CM, Firth H, Dwight T, Ercolino T, Mannelli M, Opocher G, Clifton-Bligh R, Gimm O, Maher ER, Robledo M, Gimenez-Roqueplo AP, Dahia PLM. Consensus Statement on next-generation-sequencing-based diagnostic testing of hereditary phaeochromocytomas and paragangliomas. Nat Rev Endocrinol 2017; 13:233-247. [PMID: 27857127 DOI: 10.1038/nrendo.2016.185] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are neural-crest-derived tumours of the sympathetic or parasympathetic nervous system that are often inherited and are genetically heterogeneous. Genetic testing is recommended for patients with these tumours and for family members of patients with hereditary forms of PPGLs. Due to the large number of susceptibility genes implicated in the diagnosis of inherited PPGLs, next-generation sequencing (NGS) technology is ideally suited for carrying out genetic screening of these individuals. This Consensus Statement, formulated by a study group comprised of experts in the field, proposes specific recommendations for the use of diagnostic NGS in hereditary PPGLs. In brief, the study group recommends target gene panels for screening of germ line DNA, technical adaptations to address different modes of disease transmission, orthogonal validation of NGS findings, standardized classification of variant pathogenicity and uniform reporting of the findings. The use of supplementary assays, to aid in the interpretation of the results, and sequencing of tumour DNA, for identification of somatic mutations, is encouraged. In addition, the study group launches an initiative to develop a gene-centric curated database of PPGL variants, with annual re-evaluation of variants of unknown significance by an expert group for purposes of reclassification and clinical guidance.
Collapse
Affiliation(s)
| | - Rodrigo A Toledo
- Division of Hematology and Medical Oncology, Department of Medicine, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, MC7880, San Antonio, Texas 78229, USA
- Spanish National Cancer Research Centre, CNIO, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Nelly Burnichon
- Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 20 Rue Leblanc, 75015 Paris, France
- INSERM, UMR970, Paris Cardiovascular Research Center (PARCC), 56 Rue Leblanc, 75015, Paris, France
| | - Alberto Cascon
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Diana E Benn
- Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, St Leonards, University of Sydney, Reserve Road, St Leonards, Sydney, New South Wales 2065, Australia
| | - Jean-Pierre Bayley
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, Netherlands
| | - Jenny Welander
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - Carli M Tops
- Department of Clinical Genetics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, Netherlands
| | - Helen Firth
- Department of Medical Genetics, University of Cambridge, Cambridge and NIHR Cambridge Biomedical Research Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Trish Dwight
- Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, St Leonards, University of Sydney, Reserve Road, St Leonards, Sydney, New South Wales 2065, Australia
| | - Tonino Ercolino
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Giuseppe Opocher
- Familial Cancer Clinic, Veneto Institute of Oncology, IRCCS, Via Gattamelata, 64 Padova, Veneto 35128, Padova, Italy
| | - Roderick Clifton-Bligh
- Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, St Leonards, University of Sydney, Reserve Road, St Leonards, Sydney, New South Wales 2065, Australia
| | - Oliver Gimm
- Department of Surgery, Region Östergötland, Linköping University, 581 83 Linköping, Sweden
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge and NIHR Cambridge Biomedical Research Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Anne-Paule Gimenez-Roqueplo
- Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 20 Rue Leblanc, 75015 Paris, France
- INSERM, UMR970, Paris Cardiovascular Research Center (PARCC), 56 Rue Leblanc, 75015, Paris, France
| | - Patricia L M Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, MC7880, San Antonio, Texas 78229, USA
| |
Collapse
|
13
|
Hypertension: The role of biochemistry in the diagnosis and management. Clin Chim Acta 2016; 465:131-143. [PMID: 28007614 DOI: 10.1016/j.cca.2016.12.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 12/11/2016] [Accepted: 12/14/2016] [Indexed: 01/10/2023]
Abstract
Hypertension is defined as a persistently elevated blood pressure ≥140/90mmHg. It is an important treatable risk factor for cardiovascular disease, with a high prevalence in the general population. The most common cause, essential hypertension, is a widespread disease - however, secondary hypertension is under investigated and under diagnosed. Collectively, hypertension is referred to as a "silent killer" - frequently it displays no overt symptomatology. It is a leading risk factor for death and disability globally, with >40% of persons aged over 25 having hypertension. A vast spectrum of conditions result in hypertension spanning essential through resistant, to patients with an overt endocrine cause. A significant number of patients with hypertension have multiple cardiovascular risk factors at the time of presentation. Both routine and specialised biochemical investigations are paramount for the evaluation of these patients and their subsequent management. Biochemical testing serves to identify those hypertensive individuals who are at higher risk on the basis of evidence of dysglycaemia, dyslipidaemia, renal impairment, or target organ damage and to exclude identifiable causes of hypertension. The main target of biochemical testing is the identification of patients with a specific and treatable aetiology of hypertension. Information gleaned from biochemical investigation is used to risk stratify patients and tailor the type and intensity of subsequent management and treatment. We review the approach to the biochemical investigation of patients presenting with hypertension and propose a diagnostic algorithm for work-up.
Collapse
|
14
|
Oncogene Status of an Interdigitating Dendritic Cell Sarcoma: Recurrent Mutations in NF1, TP53, and ARID2 Shared With Melanoma. Am J Surg Pathol 2016; 40:1721-1723. [DOI: 10.1097/pas.0000000000000746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Därr R, Nambuba J, Del Rivero J, Janssen I, Merino M, Todorovic M, Balint B, Jochmanova I, Prchal JT, Lechan RM, Tischler AS, Popovic V, Miljic D, Adams KT, Prall FR, Ling A, Golomb MR, Ferguson M, Nilubol N, Chen CC, Chew E, Taïeb D, Stratakis CA, Fojo T, Yang C, Kebebew E, Zhuang Z, Pacak K. Novel insights into the polycythemia-paraganglioma-somatostatinoma syndrome. Endocr Relat Cancer 2016; 23:899-908. [PMID: 27679736 PMCID: PMC5096964 DOI: 10.1530/erc-16-0231] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Worldwide, the syndromes of paraganglioma (PGL), somatostatinoma (SOM) and early childhood polycythemia are described in only a few patients with somatic mutations in the hypoxia-inducible factor 2 alpha (HIF2A). This study provides detailed information about the clinical aspects and course of 7 patients with this syndrome and brings into perspective these experiences with the pertinent literature. Six females and one male presented at a median age of 28 years (range 11-46). Two were found to have HIF2A somatic mosaicism. No relatives were affected. All patients were diagnosed with polycythemia before age 8 and before PGL/SOM developed. PGLs were found at a median age of 17 years (range 8-38) and SOMs at 29 years (range 22-38). PGLs were multiple, recurrent and metastatic in 100, 100 and 29% of all cases, and SOMs in 40, 40 and 60%, respectively. All PGLs were primarily norepinephrine-producing. All patients had abnormal ophthalmologic findings and those with SOMs had gallbladder disease. Computed tomography (CT) and magnetic resonance imaging revealed cystic lesions at multiple sites and hemangiomas in 4 patients (57%), previously thought to be pathognomonic for von Hippel-Lindau disease. The most accurate radiopharmaceutical to detect PGL appeared to be [18F]-fluorodihydroxyphenylalanine ([18F]-FDOPA). Therefore, [18F]-FDOPA PET/CT, not [68Ga]-(DOTA)-[Tyr3]-octreotate ([68Ga]-DOTATATE) PET/CT is recommended for tumor localization and aftercare in this syndrome. The long-term prognosis of the syndrome is unknown. However, to date no deaths occurred after 6 years follow-up. Physicians should be aware of this unique syndrome and its diagnostic and therapeutic challenges.
Collapse
Affiliation(s)
- Roland Därr
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Joan Nambuba
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Jaydira Del Rivero
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Ingo Janssen
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Merino
- Laboratory of PathologyNational Institutes of Health, Bethesda, Maryland, USA
| | - Milena Todorovic
- Institute of HematologyClinical Center of Serbia and Medical Faculty University of Belgrade, Belgrade, Serbia
| | - Bela Balint
- Institute of Transfusiology and Hemobiology of Military Medical Academy and Institute for Medical ResearchUniversity of Belgrade, Belgrade, Serbia
| | - Ivana Jochmanova
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- 1st Department of Internal MedicineFaculty of Medicine, Pavol Jozef Safarik University in Kosice, Kosice, Slovakia
| | - Josef T Prchal
- Division of HematologyUniversity of Utah, Salt Lake City, Utah, USA
| | - Ronald M Lechan
- Tupper Research Institute and Department of MedicineDivision of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, Boston, Massachusetts, USA
| | - Arthur S Tischler
- Department of Pathology and Laboratory MedicineTufts Medical Center, Boston, Massachusetts, USA
| | - Vera Popovic
- Institute of EndocrinologyClinical Center of Serbia, Medical Faculty, University Belgrade, Belgrade, Serbia
| | - Dragana Miljic
- Institute of EndocrinologyClinical Center of Serbia, Medical Faculty, University Belgrade, Belgrade, Serbia
| | - Karen T Adams
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - F Ryan Prall
- Department of OphthalmologyEugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Alexander Ling
- Department of Radiology and Imaging SciencesClinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Meredith R Golomb
- Division of Child NeurologyDepartment of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael Ferguson
- Riley Hospital for Children at Indiana University HealthIndianapolis, Indiana, USA
| | - Naris Nilubol
- Endocrine Oncology BranchNational Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Clara C Chen
- Division of Nuclear MedicineDepartment of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Emily Chew
- Division of Epidemiology and Clinical ApplicationsNational Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David Taïeb
- Department of Nuclear MedicineLa Timone University Hospital & CERIMED & Inserm UMR1068 Marseille Cancerology Research Center, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Constantine A Stratakis
- Division of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Tito Fojo
- Medical Oncology BranchNational Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chunzhang Yang
- Neuro-Oncology BranchCenter for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Electron Kebebew
- Endocrine Oncology BranchNational Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhengping Zhuang
- Neuro-Oncology BranchCenter for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Karel Pacak
- Section on Medical NeuroendocrinologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
|
17
|
Crona J, Skogseid B. GEP- NETS UPDATE: Genetics of neuroendocrine tumors. Eur J Endocrinol 2016; 174:R275-90. [PMID: 27165966 DOI: 10.1530/eje-15-0972] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022]
Abstract
Neuroendocrine tumors (NETs) are a heterogeneous group of neoplasms, arising from neuroendocrine cells that are dispersed throughout the body. Around 20% of NETs occur in the context of a genetic syndrome. Today there are at least ten recognized NET syndromes. This includes the classical syndromes: multiple endocrine neoplasias types 1 and 2, and von Hippel-Lindau and neurofibromatosis type 1. Additional susceptibility genes associated with a smaller fraction of NETs have also been identified. Recognizing genetic susceptibility has proved essential both to provide genetic counseling and to give the best preventive care. In this review we will also discuss the knowledge of somatic genetic alterations in NETs. At least 24 genes have been implicated as drivers of neuroendocrine tumorigenesis, and the overall rates of genomic instability are relatively low. Genetic intra-tumoral, as well as inter-tumoral heterogeneity in the same patient, have also been identified. Together these data point towards the common pathways in NET evolution, separating early from late disease drivers. Although knowledge of specific mutations in NETs has limited impact on actual patient management, we predict that in the near future genomic profiling of tumors will be included in the clinical arsenal for diagnostics, prognostics and therapeutic decisions.
Collapse
Affiliation(s)
- Joakim Crona
- Department of Medical SciencesUppsala University, Rudbecklaboratoriet, Dag hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - Britt Skogseid
- Department of Medical SciencesUppsala University, Rudbecklaboratoriet, Dag hammarskjölds väg 20, 75185 Uppsala, Sweden
| |
Collapse
|
18
|
Patócs A, Lendvai NK, Butz H, Liko I, Sapi Z, Szucs N, Toth G, Grolmusz VK, Igaz P, Toth M, Rácz K. Novel SDHB and TMEM127 Mutations in Patients with Pheochromocytoma/Paraganglioma Syndrome. Pathol Oncol Res 2016; 22:673-9. [DOI: 10.1007/s12253-016-0050-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/29/2016] [Indexed: 11/30/2022]
|
19
|
|
20
|
K.M. Ip C, Yin J, K.S. Ng P, Lin SY, B. Mills G. Genomic-Glycosylation Aberrations in Tumor Initiation, Progression and Management. AIMS MEDICAL SCIENCE 2016. [DOI: 10.3934/medsci.2016.4.386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
21
|
Toledo RA, Dahia PL. Next-generation sequencing for the diagnosis of hereditary pheochromocytoma and paraganglioma syndromes. Curr Opin Endocrinol Diabetes Obes 2015; 22:169-79. [PMID: 25871962 PMCID: PMC7216557 DOI: 10.1097/med.0000000000000150] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW About 40% of the neuroendocrine tumors pheochromocytomas and paragangliomas (PPGLs) are caused by an inherited mutation. Diagnostic genetic screening is recommended for patients and their families. However, the number of susceptibility genes involved is high and continues to grow, making conventional sequencing costly and burdensome. Next-generation sequencing (NGS) enables accurate, thorough, and cost-effective identification of inherited mutations. Here we review recent successes, limitations, and the future of NGS for diagnosis of pheochromocytoma and paraganglioma syndromes. RECENT FINDINGS NGS-based screen of genetic disorders in the clinical setting shows improved diagnostic rates over conventional tests. Both broad, whole-exome sequencing, and targeted NGS approaches have been tested for screening of PPGLs, with accurate mutation detection, higher speed, and reduced costs compared with current assays. Flexibility to expand the targeted gene set is immediate in whole-exome sequencing, and adjustable in targeted NGS, but both methods have limitations. SUMMARY The high degree of genetic heterogeneity and heritability of PPGLs make NGS an ideal medium for their diagnostic screening. However, improved detection of large genomic defects and underrepresented gene areas are needed before NGS can fully realize its potential as the premier option for routine genetic testing of these syndromes.
Collapse
Affiliation(s)
- Rodrigo A. Toledo
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Patricia L.M. Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
- Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, Texas, USA
| |
Collapse
|
22
|
SDHB/SDHA immunohistochemistry in pheochromocytomas and paragangliomas: a multicenter interobserver variation analysis using virtual microscopy: a Multinational Study of the European Network for the Study of Adrenal Tumors (ENS@T). Mod Pathol 2015; 28:807-21. [PMID: 25720320 DOI: 10.1038/modpathol.2015.41] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 01/10/2015] [Accepted: 01/10/2015] [Indexed: 12/13/2022]
Abstract
Despite the established role of SDHB/SDHA immunohistochemistry as a valuable tool to identify patients at risk for familial succinate dehydrogenase-related pheochromocytoma/paraganglioma syndromes, the reproducibility of the assessment methods has not as yet been determined. The aim of this study was to investigate interobserver variability among seven expert endocrine pathologists using a web-based virtual microscopy approach in a large multicenter pheochromocytoma/paraganglioma cohort (n=351): (1) 73 SDH mutated, (2) 105 non-SDH mutated, (3) 128 samples without identified SDH-x mutations, and (4) 45 with incomplete SDH molecular genetic analysis. Substantial agreement among all the reviewers was observed either with a two-tiered classification (SDHB κ=0.7338; SDHA κ=0.6707) or a three-tiered classification approach (SDHB κ=0.6543; SDHA κ=0.7516). Consensus was achieved in 315 cases (89.74%) for SDHB immunohistochemistry and in 348 cases (99.15%) for SDHA immunohistochemistry. Among the concordant cases, 62 of 69 (~90%) SDHB-/C-/D-/AF2-mutated cases displayed SDHB immunonegativity and SDHA immunopositivity, 3 of 4 (75%) with SDHA mutations showed loss of SDHA/SDHB protein expression, whereas 98 of 105 (93%) non-SDH-x-mutated counterparts demonstrated retention of SDHA/SDHB protein expression. Two SDHD-mutated extra-adrenal paragangliomas were scored as SDHB immunopositive, whereas 9 of 128 (7%) tumors without identified SDH-x mutations, 6 of 37 (~16%) VHL-mutated, as well as 1 of 21 (~5%) NF1-mutated tumors were evaluated as SDHB immunonegative. Although 14 out of those 16 SDHB-immunonegative cases were nonmetastatic, an overall significant correlation between SDHB immunonegativity and malignancy was observed (P=0.00019). We conclude that SDHB/SDHA immunohistochemistry is a reliable tool to identify patients with SDH-x mutations with an additional value in the assessment of genetic variants of unknown significance. If SDH molecular genetic analysis fails to detect a mutation in SDHB-immunonegative tumor, SDHC promoter methylation and/or VHL/NF1 testing with the use of targeted next-generation sequencing is advisable.
Collapse
|
23
|
Crona J, Backman S, Maharjan R, Mayrhofer M, Stålberg P, Isaksson A, Hellman P, Björklund P. Spatiotemporal Heterogeneity Characterizes the Genetic Landscape of Pheochromocytoma and Defines Early Events in Tumorigenesis. Clin Cancer Res 2015; 21:4451-60. [PMID: 25991818 DOI: 10.1158/1078-0432.ccr-14-2854] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 05/11/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Pheochromocytoma and paraganglioma (PPGL) patients display heterogeneity in the clinical presentation and underlying genetic cause. The degree of inter- and intratumor genetic heterogeneity has not yet been defined. EXPERIMENTAL DESIGN In PPGLs from 94 patients, we analyzed LOH, copy-number variations, and mutation status of SDHA, SDHB, SDHC, SDHD, SDHAF2, VHL, EPAS1, NF1, RET, TMEM127, MAX, and HRAS using high-density SNP array and targeted deep sequencing, respectively. Genetic heterogeneity was determined through (i) bioinformatics analysis of individual samples that estimated absolute purity and ploidy from SNP array data and (ii) comparison of paired tumor samples that allowed reconstruction of phylogenetic trees. RESULTS Mutations were found in 61% of the tumors and correlated with specific patterns of somatic copy-number aberrations (SCNA) and degree of nontumoral cell admixture. Intratumor genetic heterogeneity was observed in 74 of 136 samples using absolute bioinformatics estimations and in 22 of 24 patients by comparison of paired samples. In addition, a low genetic concordance was observed between paired primary tumors and distant metastases. This allowed for reconstructing the life history of individual tumors, identifying somatic mutations as well as copy-number loss of 3p and 11p (VHL subgroup), 1p (Cluster 2), and 17q (NF1 subgroup) as early events in PPGL tumorigenesis. CONCLUSIONS Genomic landscapes of PPGL are specific to mutation subtype and characterized by genetic heterogeneity both within and between tumor lesions of the same patient.
Collapse
Affiliation(s)
- Joakim Crona
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Samuel Backman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rajani Maharjan
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Markus Mayrhofer
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Isaksson
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Peyman Björklund
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
24
|
Favier J, Amar L, Gimenez-Roqueplo AP. Paraganglioma and phaeochromocytoma: from genetics to personalized medicine. Nat Rev Endocrinol 2015; 11:101-11. [PMID: 25385035 DOI: 10.1038/nrendo.2014.188] [Citation(s) in RCA: 326] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Paragangliomas and phaeochromocytomas are neuroendocrine tumours whose pathogenesis and progression are very strongly influenced by genetics. A germline mutation in one of the susceptibility genes identified so far explains ∼40% of all cases; the remaining 60% are thought to be sporadic cases. At least one-third of these sporadic tumours contain a somatic mutation in a predisposing gene. Genetic testing, which is indicated in every patient, is guided by the clinical presentation as well as by the secretory phenotype and the immunohistochemical characterization of the tumours. The diagnosis of an inherited form drives clinical management and tumour surveillance. Different 'omics' profiling methods have provided a neat classification of these tumours in accordance with their genetic background. Transcriptomic studies have identified two main molecular pathways that underlie development of these tumours, one in which the hypoxic pathway is activated (cluster 1) and another in which the MAPK and mTOR (mammalian target of rapamycin) signalling pathways are activated (cluster 2). DNA methylation profiling has uncovered a hypermethylator phenotype in tumours related to SDHx genes (a group of genes comprising SDHA, SDHB, SDHC, SDHD and SDHAF2) and revealed that succinate acts as an oncometabolite, inhibiting 2-oxoglutarate-dependent dioxygenases, such as hypoxia-inducible factor prolyl-hydroxylases and histone and DNA demethylases. 'Omics' data have suggested new therapeutic targets for patients with a malignant tumour. In the near future, new 'omics'-based tests are likely to be transferred into clinical practice with the goal of establishing personalized medical management for affected patients.
Collapse
Affiliation(s)
- Judith Favier
- INSERM, UMR 970, Paris Cardiovascular Research Centre, F-75015 Paris, France
| | - Laurence Amar
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité d'Hypertension Artérielle, F-75015 Paris, France
| | | |
Collapse
|
25
|
Canu L, Rapizzi E, Zampetti B, Fucci R, Nesi G, Richter S, Qin N, Giachè V, Bergamini C, Parenti G, Valeri A, Ercolino T, Eisenhofer G, Mannelli M. Pitfalls in genetic analysis of pheochromocytomas/paragangliomas-case report. J Clin Endocrinol Metab 2014; 99:2321-6. [PMID: 24758185 DOI: 10.1210/jc.2013-4453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT About 35% of patients with pheochromocytoma/paraganglioma carry a germline mutation in one of the 10 main susceptibility genes. The recent introduction of next-generation sequencing will allow the analysis of all these genes in one run. When positive, the analysis is generally unequivocal due to the association between a germline mutation and a concordant clinical presentation or positive family history. When genetic analysis reveals a novel mutation with no clinical correlates, particularly in the presence of a missense variant, the question arises whether the mutation is pathogenic or a rare polymorphism. OBJECTIVE We report the case of a 35-year-old patient operated for a pheochromocytoma who turned out to be a carrier of a novel SDHD (succinate dehydrogenase subunit D) missense mutation. With no positive family history or clinical correlates, we decided to perform additional analyses to test the clinical significance of the mutation. METHODS We performed in silico analysis, tissue loss of heterozygosity analysis, immunohistochemistry, Western blot analysis, SDH enzymatic assay, and measurement of the succinate/fumarate concentration ratio in the tumor tissue by tandem mass spectrometry. RESULTS Although the in silico analysis gave contradictory results according to the different methods, all the other tests demonstrated that the SDH complex was conserved and normally active. We therefore came to the conclusion that the variant was a nonpathogenic polymorphism. CONCLUSIONS Advancements in technology facilitate genetic analysis of patients with pheochromocytoma but also offer new challenges to the clinician who, in some cases, needs clinical correlates and/or functional tests to give significance to the results of the genetic assay.
Collapse
Affiliation(s)
- Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences (L.C., E.R., B.Z., R.F., V.G., M.M.), and Department of Human Pathology and Oncology (G.N.), University of Florence, 50134 Florence, Italy; General and Surgical Unit (C.B., A.V.), and Endocrinology Unit, Careggi Hospital (G.P., T.E.), Azienda Ospedaliera Universitaria Careggi, 50134 Florence, Italy; Division of Clinical Neurochemistry (S.R., N.Q., G.E.), Institute of Clinical Chemistry and Laboratory Medicine and Department of Medicine, Dresden University of Technology, 01069 Dresden, Germany; and Istituto Toscano Tumori (M.M.), 50139 Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Casey R, Garrahy A, Tuthill A, O'Halloran D, Joyce C, Casey MB, O'Shea P, Bell M. Universal genetic screening uncovers a novel presentation of an SDHAF2 mutation. J Clin Endocrinol Metab 2014; 99:E1392-6. [PMID: 24712571 DOI: 10.1210/jc.2013-4536] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
CONTEXT Hereditary pheochromocytoma/paraganglioma (PC/PGL) accounts for up to 60% of previously considered sporadic tumors. Guidelines suggest that phenotype should guide genetic testing. Next-generation sequencing technology can simultaneously sequence 9 of the 18 known susceptibility genes in a timely, cost-efficient manner. OBJECTIVE Our aim was to confirm that universal screening is superior to targeted testing in patients with histologically confirmed PC and PGL. METHODS In two tertiary referral hospitals in Ireland, NGS was carried out on all histologically confirmed cases of PC/PGL diagnosed between 2004 and 2013. The following susceptibility genes were sequenced: VHL, RET, SDHA, SDHB, SDHC, SDHD, SDHAF2, TMEM127, and MAX. A multiplex ligation-dependent probe amplification analysis was performed in VHL, SDHB, SDHC, SDHD, and SDHAF2 genes to detect deletions and duplications. RESULTS A total of 31 patients were tested, 31% (n = 10) of whom were found to have a genetic mutation. Of those patients with a positive genotype, phenotype predicted genotype in only 50% (n = 5). Significant genetic mutations that would have been missed in our cohort by phenotypic evaluation alone include a mutation in TMEM127, two mutations in SDHAF2, and two mutations in RET. Target testing would have identified three of the latter mutations based on age criteria. However, 20% of patients (n = 2) would not have satisfied any criteria for targeted testing including one patient with a novel presentation of an SDHAF2 mutation. CONCLUSION This study supports the value of universal genetic screening for all patients with PC/PGL.
Collapse
Affiliation(s)
- Ruth Casey
- Departments of Endocrinology (R.C., A.G., M.B.), Pathology (M.B.C.), and Clinical Biochemistry (P.O.), Galway University Hospital, Galway, Ireland; and Departments of Endocrinology (A.T., D.O.) and Clinical Biochemistry (C.J.), Cork University Hospital, Cork, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Welander J, Andreasson A, Juhlin CC, Wiseman RW, Bäckdahl M, Höög A, Larsson C, Gimm O, Söderkvist P. Rare germline mutations identified by targeted next-generation sequencing of susceptibility genes in pheochromocytoma and paraganglioma. J Clin Endocrinol Metab 2014; 99:E1352-60. [PMID: 24694336 PMCID: PMC5393486 DOI: 10.1210/jc.2013-4375] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/26/2014] [Indexed: 02/08/2023]
Abstract
CONTEXT Pheochromocytomas and paragangliomas have a highly diverse genetic background, with a third of the cases carrying a germline mutation in 1 of 14 identified genes. OBJECTIVE This study aimed to evaluate next-generation sequencing for more efficient genetic testing of pheochromocytoma and paraganglioma and to establish germline and somatic mutation frequencies for all known susceptibility genes. DESIGN A targeted next-generation sequencing approach on an Illumina MiSeq instrument was used for a mutation analysis in 86 unselected pheochromocytoma and paraganglioma tumor samples. The study included the genes EGLN1, EPAS1, KIF1Bβ, MAX, MEN1, NF1, RET, SDHA, SDHB, SDHC, SDHD, SDHAF2, TMEM127, and VHL. RESULTS were verified in tumor and constitutional DNA with Sanger sequencing. RESULTS In all cases with clinical syndromes or known germline mutations, a mutation was detected in the expected gene. Among 68 nonfamilial tumors, 32 mutations were identified in 28 of the samples (41%), including germline mutations in EGLN1, KIF1Bβ, SDHA, SDHB, and TMEM127 and somatic mutations in EPAS1, KIF1Bβ, MAX, NF1, RET, and VHL, including one double monoallelic EPAS1 mutation. CONCLUSIONS Targeted next-generation sequencing proved to be fast and cost effective for the genetic analysis of pheochromocytoma and paraganglioma. More than half of the tumors harbored mutations in the investigated genes. Notably, 7% of the apparently sporadic cases carried germline mutations, highlighting the importance of comprehensive genetic testing. KIF1Bβ, which previously has not been investigated in a large cohort, appears to be an equally important tumor suppressor as MAX and TMEM127 and could be considered for genetic testing of these patients.
Collapse
Affiliation(s)
| | | | - C. Christofer Juhlin
- Department of Clinical and Experimental Medicine (J.W., O.G., P.S.), Faculty of Health Sciences, Linköping University, Departments of Surgery (O.G.) and Clinical Genetics (P.S.), County Council of Östergötland, Linköping SE-58185, Sweden; Department of Oncology-Pathology (A.A., C.C.J., A.H., C.L.), Karolinska Institutet, Cancer Center Karolinska (A.A., C.C.J., A.H., C.L.), Karolinska University Hospital Solna, and Department of Molecular Medicine and Surgery (A.A., C.C.J., M.B., C.L.), Karolinska Institutet, Karolinska University Hospital, Stockholm SE-17176, Sweden; and Wisconsin National Primate Research Center (R.W.W.), University of Wisconsin-Madison, Madison, Wisconsin 53715
| | - Roger W. Wiseman
- Department of Clinical and Experimental Medicine (J.W., O.G., P.S.), Faculty of Health Sciences, Linköping University, Departments of Surgery (O.G.) and Clinical Genetics (P.S.), County Council of Östergötland, Linköping SE-58185, Sweden; Department of Oncology-Pathology (A.A., C.C.J., A.H., C.L.), Karolinska Institutet, Cancer Center Karolinska (A.A., C.C.J., A.H., C.L.), Karolinska University Hospital Solna, and Department of Molecular Medicine and Surgery (A.A., C.C.J., M.B., C.L.), Karolinska Institutet, Karolinska University Hospital, Stockholm SE-17176, Sweden; and Wisconsin National Primate Research Center (R.W.W.), University of Wisconsin-Madison, Madison, Wisconsin 53715
| | - Martin Bäckdahl
- Department of Clinical and Experimental Medicine (J.W., O.G., P.S.), Faculty of Health Sciences, Linköping University, Departments of Surgery (O.G.) and Clinical Genetics (P.S.), County Council of Östergötland, Linköping SE-58185, Sweden; Department of Oncology-Pathology (A.A., C.C.J., A.H., C.L.), Karolinska Institutet, Cancer Center Karolinska (A.A., C.C.J., A.H., C.L.), Karolinska University Hospital Solna, and Department of Molecular Medicine and Surgery (A.A., C.C.J., M.B., C.L.), Karolinska Institutet, Karolinska University Hospital, Stockholm SE-17176, Sweden; and Wisconsin National Primate Research Center (R.W.W.), University of Wisconsin-Madison, Madison, Wisconsin 53715
| | - Anders Höög
- Department of Clinical and Experimental Medicine (J.W., O.G., P.S.), Faculty of Health Sciences, Linköping University, Departments of Surgery (O.G.) and Clinical Genetics (P.S.), County Council of Östergötland, Linköping SE-58185, Sweden; Department of Oncology-Pathology (A.A., C.C.J., A.H., C.L.), Karolinska Institutet, Cancer Center Karolinska (A.A., C.C.J., A.H., C.L.), Karolinska University Hospital Solna, and Department of Molecular Medicine and Surgery (A.A., C.C.J., M.B., C.L.), Karolinska Institutet, Karolinska University Hospital, Stockholm SE-17176, Sweden; and Wisconsin National Primate Research Center (R.W.W.), University of Wisconsin-Madison, Madison, Wisconsin 53715
| | - Catharina Larsson
- Department of Clinical and Experimental Medicine (J.W., O.G., P.S.), Faculty of Health Sciences, Linköping University, Departments of Surgery (O.G.) and Clinical Genetics (P.S.), County Council of Östergötland, Linköping SE-58185, Sweden; Department of Oncology-Pathology (A.A., C.C.J., A.H., C.L.), Karolinska Institutet, Cancer Center Karolinska (A.A., C.C.J., A.H., C.L.), Karolinska University Hospital Solna, and Department of Molecular Medicine and Surgery (A.A., C.C.J., M.B., C.L.), Karolinska Institutet, Karolinska University Hospital, Stockholm SE-17176, Sweden; and Wisconsin National Primate Research Center (R.W.W.), University of Wisconsin-Madison, Madison, Wisconsin 53715
| | | | | |
Collapse
|
28
|
Onsongo G, Erdmann J, Spears MD, Chilton J, Beckman KB, Hauge A, Yohe S, Schomaker M, Bower M, Silverstein KAT, Thyagarajan B. Implementation of Cloud based next generation sequencing data analysis in a clinical laboratory. BMC Res Notes 2014; 7:314. [PMID: 24885806 PMCID: PMC4036707 DOI: 10.1186/1756-0500-7-314] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 05/06/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The introduction of next generation sequencing (NGS) has revolutionized molecular diagnostics, though several challenges remain limiting the widespread adoption of NGS testing into clinical practice. One such difficulty includes the development of a robust bioinformatics pipeline that can handle the volume of data generated by high-throughput sequencing in a cost-effective manner. Analysis of sequencing data typically requires a substantial level of computing power that is often cost-prohibitive to most clinical diagnostics laboratories. FINDINGS To address this challenge, our institution has developed a Galaxy-based data analysis pipeline which relies on a web-based, cloud-computing infrastructure to process NGS data and identify genetic variants. It provides additional flexibility, needed to control storage costs, resulting in a pipeline that is cost-effective on a per-sample basis. It does not require the usage of EBS disk to run a sample. CONCLUSIONS We demonstrate the validation and feasibility of implementing this bioinformatics pipeline in a molecular diagnostics laboratory. Four samples were analyzed in duplicate pairs and showed 100% concordance in mutations identified. This pipeline is currently being used in the clinic and all identified pathogenic variants confirmed using Sanger sequencing further validating the software.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kevin A T Silverstein
- Research Informatics Support Systems, Minnesota Supercomputing Institute, University of Minnesota, Room 599 Walter Library 117 Pleasant St SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
29
|
Crona J, Nordling M, Maharjan R, Granberg D, Stålberg P, Hellman P, Björklund P. Integrative genetic characterization and phenotype correlations in pheochromocytoma and paraganglioma tumours. PLoS One 2014; 9:e86756. [PMID: 24466223 PMCID: PMC3899286 DOI: 10.1371/journal.pone.0086756] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/13/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND About 60% of Pheochromocytoma (PCC) and Paraganglioma (PGL) patients have either germline or somatic mutations in one of the 12 proposed disease causing genes; SDHA, SDHB, SDHC, SDHD, SDHAF2, VHL, EPAS1, RET, NF1, TMEM127, MAX and H-RAS. Selective screening for germline mutations is routinely performed in clinical management of these diseases. Testing for somatic alterations is not performed on a regular basis because of limitations in interpreting the results. AIM The purpose of the study was to investigate genetic events and phenotype correlations in a large cohort of PCC and PGL tumours. METHODS A total of 101 tumours from 89 patients with PCC and PGL were re-sequenced for a panel of 10 disease causing genes using automated Sanger sequencing. Selected samples were analysed with Multiplex Ligation-dependent Probe Amplification and/or SNParray. RESULTS Pathogenic genetic variants were found in tumours from 33 individual patients (37%), 14 (16%) were discovered in constitutional DNA and 16 (18%) were confirmed as somatic. Loss of heterozygosity (LOH) was observed in 1/1 SDHB, 11/11 VHL and 3/3 NF1-associated tumours. In patients with somatic mutations there were no recurrences in contrast to carriers of germline mutations (P = 0.022). SDHx/VHL/EPAS1 associated cases had higher norepinephrine output (P = 0.03) and lower epinephrine output (P<0.001) compared to RET/NF1/H-RAS cases. CONCLUSION Somatic mutations are frequent events in PCC and PGL tumours. Tumour genotype may be further investigated as prognostic factors in these diseases. Growing evidence suggest that analysis of tumour DNA could have an impact on the management of these patients.
Collapse
Affiliation(s)
- Joakim Crona
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Margareta Nordling
- Department of Clinical Genetics, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Rajani Maharjan
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Dan Granberg
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Peyman Björklund
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
30
|
Toledo RA, Dahia PLM. Next-generation sequencing for the genetic screening of phaeochromcytomas and paragangliomas: riding the new wave, but with caution. Clin Endocrinol (Oxf) 2014; 80:23-4. [PMID: 24168015 DOI: 10.1111/cen.12357] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Rodrigo A Toledo
- Division of Hematology and Medical Oncology, Department of Medicine, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
31
|
McInerney-Leo AM, Marshall MS, Gardiner B, Benn DE, McFarlane J, Robinson BG, Brown MA, Leo PJ, Clifton-Bligh RJ, Duncan EL. Whole exome sequencing is an efficient and sensitive method for detection of germline mutations in patients with phaeochromcytomas and paragangliomas. Clin Endocrinol (Oxf) 2014; 80:25-33. [PMID: 24102379 DOI: 10.1111/cen.12331] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 08/08/2013] [Accepted: 09/12/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Genetic testing is recommended when the probability of a disease-associated germline mutation exceeds 10%. Germline mutations are found in approximately 25% of individuals with phaeochromcytoma (PCC) or paraganglioma (PGL); however, genetic heterogeneity for PCC/PGL means many genes may require sequencing. A phenotype-directed iterative approach may limit costs but may also delay diagnosis, and will not detect mutations in genes not previously associated with PCC/PGL. OBJECTIVE To assess whether whole exome sequencing (WES) was efficient and sensitive for mutation detection in PCC/PGL. METHODS Whole exome sequencing was performed on blinded samples from eleven individuals with PCC/PGL and known mutations. Illumina TruSeq (Illumina Inc, San Diego, CA, USA) was used for exome capture of seven samples, and NimbleGen SeqCap EZ v3.0 (Roche NimbleGen Inc, Basel, Switzerland) for five samples (one sample was repeated). Massive parallel sequencing was performed on multiplexed samples. Sequencing data were called using Genome Analysis Toolkit and annotated using annovar. Data were assessed for coding variants in RET, NF1, VHL, SDHD, SDHB, SDHC, SDHA, SDHAF2, KIF1B, TMEM127, EGLN1 and MAX. Target capture of five exome capture platforms was compared. RESULTS Six of seven mutations were detected using Illumina TruSeq exome capture. All five mutations were detected using NimbleGen SeqCap EZ v3.0 platform, including the mutation missed using Illumina TruSeq capture. Target capture for exons in known PCC/PGL genes differs substantially between platforms. Exome sequencing was inexpensive (<$A800 per sample for reagents) and rapid (results <5 weeks from sample reception). CONCLUSION Whole exome sequencing is sensitive, rapid and efficient for detection of PCC/PGL germline mutations. However, capture platform selection is critical to maximize sensitivity.
Collapse
Affiliation(s)
- Aideen M McInerney-Leo
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Martins R, Bugalho MJ. Paragangliomas/Pheochromocytomas: clinically oriented genetic testing. Int J Endocrinol 2014; 2014:794187. [PMID: 24899893 PMCID: PMC4037125 DOI: 10.1155/2014/794187] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Paragangliomas are rare neuroendocrine tumors that arise in the sympathetic or parasympathetic nervous system. Sympathetic paragangliomas are mainly found in the adrenal medulla (designated pheochromocytomas) but may also have a thoracic, abdominal, or pelvic localization. Parasympathetic paragangliomas are generally located at the head or neck. Knowledge concerning the familial forms of paragangliomas has greatly improved in recent years. Additionally to the genes involved in the classical syndromic forms: VHL gene (von Hippel-Lindau), RET gene (Multiple Endocrine Neoplasia type 2), and NF1 gene (Neurofibromatosis type 1), 10 novel genes have so far been implicated in the occurrence of paragangliomas/pheochromocytomas: SDHA, SDHB, SDHC, SDHD, SDHAF2, TMEM127, MAX, EGLN1, HIF2A, and KIF1B. It is currently accepted that about 35% of the paragangliomas cases are due to germline mutations in one of these genes. Furthermore, somatic mutations of RET, VHL, NF1, MAX, HIF2A, and H-RAS can also be detected. The identification of the mutation responsible for the paraganglioma/pheochromocytoma phenotype in a patient may be crucial in determining the treatment and allowing specific follow-up guidelines, ultimately leading to a better prognosis. Herein, we summarize the most relevant aspects regarding the genetics and clinical aspects of the syndromic and nonsyndromic forms of pheochromocytoma/paraganglioma aiming to provide an algorithm for genetic testing.
Collapse
Affiliation(s)
- Rute Martins
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Maria João Bugalho
- Serviço de Endocrinologia, Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., 1099-023 Lisboa, Portugal
- Clínica Universitária de Endocrinologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
- *Maria João Bugalho:
| |
Collapse
|
33
|
MAX mutations status in Swedish patients with pheochromocytoma and paraganglioma tumours. Fam Cancer 2013; 13:121-5. [DOI: 10.1007/s10689-013-9666-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|