1
|
Giacché M, Tacchetti MC, Agabiti-Rosei C, Torlone F, Bandera F, Izzi C, Agabiti-Rosei E. Pheochromocytoma-Paraganglioma Syndrome: A Multiform Disease with Different Genotype and Phenotype Features. Biomedicines 2024; 12:2385. [PMID: 39457697 PMCID: PMC11504466 DOI: 10.3390/biomedicines12102385] [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: 09/07/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pheochromocytoma and paraganglioma (PPGL) are rare tumors derived from the adrenal medulla and extra-adrenal chromaffin cells. Diagnosis is often challenging due to the great variability in clinical presentation; the complexity of management due to the dangerous effects of catecholamine excess and the potentially malignant behavior require in-depth knowledge of the pathology and multidisciplinary management. Nowadays, diagnostic ability has certainly improved and guidelines and consensus documents for treatment and follow-up are available. A major impulse to the development of this knowledge has come from the new findings on the genetic and molecular characteristics of PPGLs. Germline mutation in susceptibility genes is detected in 40% of subjects, with a mutation frequency of 10-12% also in patients with sporadic presentation and genetic testing should be incorporated within clinical care. PPGL susceptibility genes include "old genes" associated with Neurofibromatosis type 1 (NF1 gene), Von Hippel Lindau syndrome (VHL gene) and Multiple Endocrine Neoplasia type 2 syndrome (RET gene), the family of SDHx genes (SDHA, SDHB, SDHC, SDHD, SDHAF2), and genes less frequently involved such as TMEM, MAX, and FH. Each gene has a different risk of relapse, malignancy, and other organ involvement; for mutation carriers, affected or asymptomatic, it is possible to define a tailored long-life surveillance program according to the gene involved. In addition, molecular characterization of the tumor has allowed the identification of somatic mutations in other driver genes, bringing to 70% the PPGLs for which we know the mechanisms of tumorigenesis. This has expanded the catalog of tumor driver genes, which are identifiable in up to 70% of patients Integrated genomic and transcriptomic data over the last 10 years have revealed three distinct major molecular signatures, triggered by pathogenic variants in susceptibility genes and characterized by the activation of a specific oncogenic signaling: the pseudo hypoxic, the kinase, and the Wnt signaling pathways. These molecular clusters show a different biochemical phenotype and clinical behavior; they may also represent the prerequisite for implementing customized therapy and follow-up.
Collapse
Affiliation(s)
- Mara Giacché
- Department of Medical and Surgical Specialties, Division of Clinical Genetics, ASST-Spedali Civili, 25133 Brescia, Italy
| | - Maria Chiara Tacchetti
- Division Internal Medicine 2, Department of Clinical and Experimental Sciences, University of Brescia, ASST-Spedali Civili, 25133 Brescia, Italy; (M.C.T.); (C.A.-R.)
| | - Claudia Agabiti-Rosei
- Division Internal Medicine 2, Department of Clinical and Experimental Sciences, University of Brescia, ASST-Spedali Civili, 25133 Brescia, Italy; (M.C.T.); (C.A.-R.)
| | - Francesco Torlone
- Clinical Research Hospital, IRCCS Multimedica, Sesto San Giovanni, 20099 Milan, Italy; (F.T.); (F.B.)
| | - Francesco Bandera
- Clinical Research Hospital, IRCCS Multimedica, Sesto San Giovanni, 20099 Milan, Italy; (F.T.); (F.B.)
| | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia, ASST-Spedali Civili, 25133 Brescia, Italy;
| | - Enrico Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia, Clinical Research Hospital, IRCCS Multimedica, Sesto San Giovanni, 20099 Milan, Italy
| |
Collapse
|
2
|
Lussey-Lepoutre C, Pacak K, Grossman A, Taieb D, Amar L. Overview of recent guidelines and consensus statements on initial screening and management of phaeochromocytoma and paraganglioma in SDHx pathogenic variant carriers and patients. Best Pract Res Clin Endocrinol Metab 2024:101938. [PMID: 39271377 DOI: 10.1016/j.beem.2024.101938] [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] [Indexed: 09/15/2024]
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumours with a strong genetic predisposition, involving over 20 genes and with germline pathogenic variants identified in 40 % of cases. The succinate dehydrogenase (SDHx) genes are the most commonly implicated in hereditary PPGLs, accounting for 20 % of cases, and present unique diagnostic and treatment challenges due to their potential for multiple, recurrent, and aggressive manifestations, often necessitating lifelong follow-up. Over the past two decades, advances in biochemical and imaging assessments, management, and follow-up protocols have significantly improved care for both adult and paediatric patients. These advances include next-generation sequencing, new biochemical tests, cluster-specific functional imaging, and improved surgical and radiotherapy techniques, such as stereotactic surgery and peptide receptor radionuclide therapy (PRRT). International consensus guidelines have been developed to standardise the management of patients with SDHx pathogenic variants, emphasising multidisciplinary approaches and frequent tumour board discussions. These guidelines, summarised below, cover recommendations for initial genetic testing, tumour screening, follow-up care, and management of patients and asymptomatic carriers.
Collapse
Affiliation(s)
- Charlotte Lussey-Lepoutre
- Sorbonne University, Nuclear Medicine Department, Pitié-Salpêtrière Hospital, Assistance -Publique Hôpitaux de Paris, Paris Cardiovascular Research Center (PARCC), Inserm U970, Paris, France.
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, CRC, Room 1E-3140, 10 Center Drive MSC-1109, Bethesda, MD 20892-1109, USA.
| | - Ashley Grossman
- Green Templeton College, University of Oxford, UK; NET Unit, Royal Free Hospital, London, UK; Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, University of Oxford, UK.
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, Inserm ERL U1326 RNAnoTher, Aix-Marseille Univ, France.
| | - Laurence Amar
- Université Paris Cité, AP-HP, Hôpital Européen Georges Pompidou, DMU Carte, Unité Hypertension Artérielle, Centre de références en maladies rares de la surrénale, Paris Centre de Recherche Cardiovasculaire, INSERM, Paris, France.
| |
Collapse
|
3
|
Cicchetti R, Basconi M, Litterio G, Mascitti M, Tamborino F, Orsini A, Digiacomo A, Ferro M, Schips L, Marchioni M. Advances in Molecular Mechanisms of Kidney Disease: Integrating Renal Tumorigenesis of Hereditary Cancer Syndrome. Int J Mol Sci 2024; 25:9060. [PMID: 39201746 PMCID: PMC11355026 DOI: 10.3390/ijms25169060] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Renal cell carcinoma (RCC) comprises various histologically distinct subtypes, each characterized by specific genetic alterations, necessitating individualized management and treatment strategies for each subtype. An exhaustive search of the PubMed database was conducted without any filters or restrictions. Inclusion criteria encompassed original English articles focusing on molecular mechanisms of kidney cancer. On the other hand, all non-original articles and articles published in any language other than English were excluded. Hereditary kidney cancer represents 5-8% of all kidney cancer cases and is associated with syndromes such as von Hippel-Lindau syndrome, Birt-Hogg-Dubè syndrome, succinate dehydrogenase-deficient renal cell cancer syndrome, tuberous sclerosis complex, hereditary papillary renal cell carcinoma, fumarate hydratase deficiency syndrome, BAP1 tumor predisposition syndrome, and other uncommon hereditary cancer syndromes. These conditions are characterized by distinct genetic mutations and related extra-renal symptoms. The majority of renal cell carcinoma predispositions stem from loss-of-function mutations in tumor suppressor genes. These mutations promote malignant advancement through the somatic inactivation of the remaining allele. This review aims to elucidate the main molecular mechanisms underlying the pathophysiology of major syndromes associated with renal cell carcinoma. By providing a comprehensive overview, it aims to facilitate early diagnosis and to highlight the principal therapeutic options available.
Collapse
Affiliation(s)
- Rossella Cicchetti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Martina Basconi
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Giulio Litterio
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Marco Mascitti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Flavia Tamborino
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Angelo Orsini
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Alessio Digiacomo
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20141 Milan, Italy;
| | - Luigi Schips
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Michele Marchioni
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| |
Collapse
|
4
|
Prado Wohlwend S, Bello Arques P. Radio theranostics in paragangliomas and pheochromocytomas. Rev Esp Med Nucl Imagen Mol 2024; 43:500017. [PMID: 38735639 DOI: 10.1016/j.remnie.2024.500017] [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: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
This continuing education aims to present in a clear and easy-to-understand manner the biology of paragangliomas and pheochromocytomas (PPGLs), the functional imaging studies available for their diagnosis and therapeutic planning, the requirements necessary to administer radioligand therapy (RLT) and the characteristics of these treatments (inclusion criteria, administration protocols, adverse effects and future perspectives). In this pathology we have two RLT options: [131I]MIBG and [177Lu]Lu-DOTA-TATE. The indication for treatment is determined by the expression of its therapeutic target in functional imaging studies, allowing precision and personalized medicine. Although most of the results we have for both treatments have as origin small retrospective series, RLT is presented as a safe and well-tolerated therapeutic option in PPGLs with slow-moderate progression or with uncontrollable symptoms, obtaining high disease control rates.
Collapse
Affiliation(s)
- Stefan Prado Wohlwend
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Clinical Center of Excellence Pheo Para Alliance.
| | - Pilar Bello Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Clinical Center of Excellence Pheo Para Alliance
| |
Collapse
|
5
|
Torresan F, Iacobone C, Giorgino F, Iacobone M. Genetic and Molecular Biomarkers in Aggressive Pheochromocytomas and Paragangliomas. Int J Mol Sci 2024; 25:7142. [PMID: 39000254 PMCID: PMC11241596 DOI: 10.3390/ijms25137142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neoplasms producing catecholamines that occur as hereditary syndromes in 25-40% of cases. To date, PPGLs are no longer classified as benign and malignant tumors since any lesion could theoretically metastasize, even if it occurs only in a minority of cases (approximately 10-30%). Over the last decades, several attempts were made to develop a scoring system able to predict the risk of aggressive behavior at diagnosis, including the risk of metastases and disease recurrence; unfortunately, none of the available scores is able to accurately predict the risk of aggressive behavior, even including clinical, biochemical, and histopathological features. Thus, life-long follow-up is required in PPGL patients. Some recent studies focusing on genetic and molecular markers (involved in hypoxia regulation, gene transcription, cellular growth, differentiation, signaling pathways, and apoptosis) seem to indicate they are promising prognostic factors, even though their clinical significance needs to be further evaluated. The most involved pathways in PPGLs with aggressive behavior are represented by Krebs cycle alterations caused by succinate dehydrogenase subunits (SDHx), especially when caused by SDHB mutations, and by fumarate hydratase mutations that lead to the activation of hypoxia pathways and DNA hypermethylation, suggesting a common pathway in tumorigenesis. Conversely, PPGLs showing mutations in the kinase cascade (cluster 2) tend to display less aggressive behavior. Finally, establishing pathways of tumorigenesis is also fundamental to developing new drugs targeted to specific pathways and improving the survival of patients with metastatic disease. Unfortunately, the rarity of these tumors and the scarce number of cases enrolled in the available studies represents an obstacle to validating the role of molecular markers as reliable predictors of aggressiveness.
Collapse
Affiliation(s)
- Francesca Torresan
- Endocrine Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy;
| | - Clelia Iacobone
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70121 Bari, Italy; (C.I.); (F.G.)
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70121 Bari, Italy; (C.I.); (F.G.)
| | - Maurizio Iacobone
- Endocrine Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy;
| |
Collapse
|
6
|
Mustafa K, Zadeh S, Culver SA. Dopamine-Secreting Carotid Body Paraganglioma in a Patient With SDHB Mutation. AACE Clin Case Rep 2024; 10:109-112. [PMID: 38799041 PMCID: PMC11127581 DOI: 10.1016/j.aace.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 05/29/2024] Open
Abstract
Background/Objective Exclusively dopamine-secreting paragangliomas (PGLs) are rare, and the majority of head and neck PGLs are nonsecretory. Here, we describe a patient with succinate dehydrogenase subunit B (SDHB) mutation and a dopamine-secreting carotid body PGL to highlight the potential importance of screening for dopamine excess in patients with suspected PGL. Case Report We report a 34-year-old patient who presented with cranial nerve palsy and was found to have a cerebellopontine PGL. Biochemical testing demonstrated increased circulating dopamine levels with normal levels of other catecholamines. Dopamine excess improved with resection of the PGL, and subsequent genetic testing revealed an SDHB mutation. Discussion Secretory head and neck PGLs and exclusively dopamine-secreting PGLs are both uncommon and rarely present together, although PGLs in patients with SDHB mutations often do produce dopamine. Although current guidelines do not recommend routine evaluation of the dopamine levels in patients at risk for PGL, dopamine-secreting PGLs are frequently locally invasive or metastatic. Conclusion Screening for dopamine excess in patients with a predisposition to PGL or with suspected PGL may aid in diagnosis and as a marker of successful treatment.
Collapse
Affiliation(s)
- Katherine Mustafa
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Sara Zadeh
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia
| | - Silas A. Culver
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
7
|
Taïeb D, Nölting S, Perrier ND, Fassnacht M, Carrasquillo JA, Grossman AB, Clifton-Bligh R, Wanna GB, Schwam ZG, Amar L, Bourdeau I, Casey RT, Crona J, Deal CL, Del Rivero J, Duh QY, Eisenhofer G, Fojo T, Ghayee HK, Gimenez-Roqueplo AP, Gill AJ, Hicks R, Imperiale A, Jha A, Kerstens MN, de Krijger RR, Lacroix A, Lazurova I, Lin FI, Lussey-Lepoutre C, Maher ER, Mete O, Naruse M, Nilubol N, Robledo M, Sebag F, Shah NS, Tanabe A, Thompson GB, Timmers HJLM, Widimsky J, Young WJ, Meuter L, Lenders JWM, Pacak K. Management of phaeochromocytoma and paraganglioma in patients with germline SDHB pathogenic variants: an international expert Consensus statement. Nat Rev Endocrinol 2024; 20:168-184. [PMID: 38097671 DOI: 10.1038/s41574-023-00926-0] [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] [Accepted: 11/16/2023] [Indexed: 02/17/2024]
Abstract
Adult and paediatric patients with pathogenic variants in the gene encoding succinate dehydrogenase (SDH) subunit B (SDHB) often have locally aggressive, recurrent or metastatic phaeochromocytomas and paragangliomas (PPGLs). Furthermore, SDHB PPGLs have the highest rates of disease-specific morbidity and mortality compared with other hereditary PPGLs. PPGLs with SDHB pathogenic variants are often less differentiated and do not produce substantial amounts of catecholamines (in some patients, they produce only dopamine) compared with other hereditary subtypes, which enables these tumours to grow subclinically for a long time. In addition, SDHB pathogenic variants support tumour growth through high levels of the oncometabolite succinate and other mechanisms related to cancer initiation and progression. As a result, pseudohypoxia and upregulation of genes related to the hypoxia signalling pathway occur, promoting the growth, migration, invasiveness and metastasis of cancer cells. These factors, along with a high rate of metastasis, support early surgical intervention and total resection of PPGLs, regardless of the tumour size. The treatment of metastases is challenging and relies on either local or systemic therapies, or sometimes both. This Consensus statement should help guide clinicians in the diagnosis and management of patients with SDHB PPGLs.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, Aix-Marseille University, La Timone University Hospital, Marseille, France
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nancy D Perrier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Jorge A Carrasquillo
- Molecular Imaging and Therapy Service, Radiology Department, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- NET Unit, Royal Free Hospital, London, UK
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital and Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - George B Wanna
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zachary G Schwam
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laurence Amar
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Hypertension Unit, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Ruth T Casey
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Joakim Crona
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cheri L Deal
- Research Center, CHU Sainte-Justine and Dept. of Paediatrics, University of Montreal, Montreal, Québec, Canada
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Quan-Yang Duh
- Department of Surgery, UCSF-Mount Zion, San Francisco, CA, USA
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Tito Fojo
- Columbia University Irving Medical Center, New York City, NY, USA
- James J. Peters VA Medical Center, New York City, NY, USA
| | - Hans K Ghayee
- Division of Endocrinology & Metabolism, Department of Medicine, University of Florida, Gainesville, FL, USA
- Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Department of Oncogenetics and Cancer Genomic Medicine, AP-HP, Hôpital européen Georges Pompidou, Paris, France
| | - Antony J Gill
- University of Sydney, Sydney NSW Australia, Cancer Diagnosis and Pathology Group Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- NSW Health Pathology Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Rodney Hicks
- Department of Medicine, St Vincent's Hospital Medical School, Melbourne, Victoria, Australia
| | - Alessio Imperiale
- Department of Nuclear Medicine and Molecular Imaging - Institut de Cancérologie de Strasbourg Europe (ICANS), IPHC, UMR 7178, CNRS, University of Strasbourg, Strasbourg, France
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michiel N Kerstens
- Department of Endocrinology, University Medical Center Groningen, Groningen, Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Máxima Center for paediatric oncology, Utrecht, Netherlands
| | - André Lacroix
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Canada
| | - Ivica Lazurova
- Department of Internal Medicine 1, University Hospital, P.J. Šafárik University, Košice, Slovakia
| | - Frank I Lin
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charlotte Lussey-Lepoutre
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Sorbonne University, Department of Nuclear Medicine, Pitié-Salpêtrière, Paris, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ozgur Mete
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mitsuhide Naruse
- Clinical Research Institute of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center and Endocrine Center, Kyoto, Japan
- Clinical Research Center, Ijinkai Takeda General Hospital, Kyoto, Japan
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Frédéric Sebag
- Department of Endocrine Surgery, Aix-Marseille University, Conception Hospital, Marseille, France
| | - Nalini S Shah
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Akiyo Tanabe
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine, Tokyo, Japan
| | - Geoffrey B Thompson
- Division of Endocrine Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jiri Widimsky
- Third Department of Medicine, Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - William J Young
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Leah Meuter
- Stanford University School of Medicine, Department of Physician Assistant Studies, Stanford, CA, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Gabiache G, Zadro C, Rozenblum L, Vezzosi D, Mouly C, Thoulouzan M, Guimbaud R, Otal P, Dierickx L, Rousseau H, Trepanier C, Dercle L, Mokrane FZ. Image-Guided Precision Medicine in the Diagnosis and Treatment of Pheochromocytomas and Paragangliomas. Cancers (Basel) 2023; 15:4666. [PMID: 37760633 PMCID: PMC10526298 DOI: 10.3390/cancers15184666] [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: 06/15/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
In this comprehensive review, we aimed to discuss the current state-of-the-art medical imaging for pheochromocytomas and paragangliomas (PPGLs) diagnosis and treatment. Despite major medical improvements, PPGLs, as with other neuroendocrine tumors (NETs), leave clinicians facing several challenges; their inherent particularities and their diagnosis and treatment pose several challenges for clinicians due to their inherent complexity, and they require management by multidisciplinary teams. The conventional concepts of medical imaging are currently undergoing a paradigm shift, thanks to developments in radiomic and metabolic imaging. However, despite active research, clinical relevance of these new parameters remains unclear, and further multicentric studies are needed in order to validate and increase widespread use and integration in clinical routine. Use of AI in PPGLs may detect changes in tumor phenotype that precede classical medical imaging biomarkers, such as shape, texture, and size. Since PPGLs are rare, slow-growing, and heterogeneous, multicentric collaboration will be necessary to have enough data in order to develop new PPGL biomarkers. In this nonsystematic review, our aim is to present an exhaustive pedagogical tool based on real-world cases, dedicated to physicians dealing with PPGLs, augmented by perspectives of artificial intelligence and big data.
Collapse
Affiliation(s)
- Gildas Gabiache
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Charline Zadro
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Laura Rozenblum
- Department of Nuclear Medicine, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 75013 Paris, France
| | - Delphine Vezzosi
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | - Céline Mouly
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | | | - Rosine Guimbaud
- Department of Oncology, Rangueil University Hospital, 31400 Toulouse, France
| | - Philippe Otal
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Lawrence Dierickx
- Department of Nuclear Medicine, IUCT-Oncopole, 31059 Toulouse, France;
| | - Hervé Rousseau
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Christopher Trepanier
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Laurent Dercle
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fatima-Zohra Mokrane
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| |
Collapse
|
9
|
Nezu M, Hirotsu Y, Amemiya K, Tateno T, Takizawa S, Inoue M, Mochizuki H, Hosaka K, Chik C, Oyama T, Omata M. Paraganglioma with High Levels of Dopamine, Dopa Decarboxylase Suppression, Dopamine β-hydroxylase Upregulation and Intra-tumoral Melanin Accumulation: A Case Report with a Literature Review. Intern Med 2023; 62:1895-1905. [PMID: 36384901 PMCID: PMC10372287 DOI: 10.2169/internalmedicine.0743-22] [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: 07/28/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Object Exclusively dopamine-producing pheochromocytoma/paraganglioma (PPGL) is an extremely rare subtype. In this condition, intratumoral dopamine β-hydroxylase (DBH), which controls the conversion of norepinephrine from dopamine, is impaired, resulting in suppressed norepinephrine and epinephrine production. However, the rarity of this type of PPGL hampers the understanding of its pathophysiology. We therefore conducted genetic and immunohistological analyses of a patient with an exclusively dopamine-producing paraganglioma. Methods Paraganglioma samples from a 52-year-old woman who presented with a 29.6- and 41.5-fold increase in plasma and 24-h urinary dopamine, respectively, but only a minor elevation in the plasma norepinephrine level was subjected to immunohistological and gene expression analyses of catecholamine synthases. Three tumors carrying known somatic PPGL-related gene variants (HRAS, EPAS1) were used as controls. Whole-exome sequencing (WES) was also performed using the patient's blood and tumor tissue. Results Surprisingly, the protein expression of DBH was not suppressed, and its mRNA expression was clearly higher in the patient than in the controls. Furthermore, dopa decarboxylase (DDC), which governs the conversion of 3,4-dihydroxyphenyl-L-alanine (L-DOPA) to dopamine, was downregulated at the protein and gene levels. In addition, melanin, which is synthesized by L-DOPA, accumulated in the tumor. WES revealed no PPGL-associated pathogenic germline variants, but a missense somatic variant (c.1798G>T) in CSDE1 was identified. Conclusion Although pre-operative plasma L-DOPA was not measured, our histological and gene expression analyses suggest that L-DOPA, rather than dopamine, might have been overproduced in the tumor. This raises the possibility of pathophysiological heterogeneity in exclusively dopamine-producing PPGL.
Collapse
Affiliation(s)
- Masahiro Nezu
- Department of Endocrinology and Diabetes, Yamanashi Central Hospital, Japan
- Genome Analysis Center, Yamanashi Central Hospital, Japan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Canada
| | - Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, Japan
| | - Kenji Amemiya
- Genome Analysis Center, Yamanashi Central Hospital, Japan
| | - Toru Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Canada
| | - Soichi Takizawa
- Department of Endocrinology and Diabetes, Yamanashi Central Hospital, Japan
| | - Masaharu Inoue
- Department of Endocrinology and Diabetes, Yamanashi Central Hospital, Japan
| | | | - Kyoko Hosaka
- Department of Urology, Yamanashi Central Hospital, Japan
| | - Constance Chik
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Canada
| | - Toshio Oyama
- Department of Pathology, Yamanashi Central Hospital, Japan
| | - Masao Omata
- Genome Analysis Center, Yamanashi Central Hospital, Japan
- The University of Tokyo, Japan
| |
Collapse
|
10
|
Lundholm MD, Marquard J, Rao PP. Paraganglioma in pregnancy, a mimic of preeclampsia: a case report. J Med Case Rep 2023; 17:124. [PMID: 37024931 PMCID: PMC10080865 DOI: 10.1186/s13256-023-03871-8] [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: 09/14/2022] [Accepted: 03/03/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND The new presentation of pheochromocytoma or paraganglioma in pregnancy is very rare and can be life-threatening for mother and child. CASE PRESENTATION We present the case of a 26-year-old gravida 3 para 2 otherwise healthy Caucasian woman at 34 weeks gestation who presented with new onset hypertension associated with headaches, dry heaves, diaphoresis, and palpitations. She was initially diagnosed with preeclampsia and treated with labetalol and an urgent cesarean section, delivering a healthy baby girl. The diagnosis of preeclampsia came into question when, 6 weeks postpartum, she continued to have hypertension with atypical features. Testing revealed metastatic paraganglioma associated with a succinate dehydrogenase B gene mutation. The patient was then started on alpha-adrenergic blockade and has had close blood pressure monitoring while discussion of advances therapies is ongoing. CONCLUSION This case demonstrates how paraganglioma/pheochromocytoma can be misdiagnosed as preeclampsia due to the overlapping features of new-onset hypertension late in pregnancy accompanied by headache and proteinuria. It is impractical to routinely screen for paraganglioma/pheochromocytoma in all pregnant patients diagnosed with preeclampsia due to the rarity of these tumors and the harm from high false-positive rates. Therefore, it is incumbent on the provider to have a high degree of suspicion for paraganglioma/pheochromocytoma when clinical features are unusual for preeclampsia, such as intermittent palpitations, diaphoresis, orthostatic hypotension, or hyperglycemia. Early detection of paraganglioma/pheochromocytoma with interventions to mitigate the risk of hypertensive crisis greatly reduce maternal and fetal mortality. Fortunately, our patient delivered a healthy baby and did not have any additional pregnancy complications despite the delay in her diagnosis.
Collapse
Affiliation(s)
- Michelle D Lundholm
- Department of Endocrinology, Diabetes and Metabolism Cleveland Clinic, 9500 Euclid Avenue, F20, Cleveland, OH, 44195, USA.
| | - Jessica Marquard
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Pratibha Pr Rao
- Department of Endocrinology, Diabetes and Metabolism Cleveland Clinic, 9500 Euclid Avenue, F20, Cleveland, OH, 44195, USA
| |
Collapse
|
11
|
Araujo-Castro M, García Sanz I, Mínguez Ojeda C, Calatayud M, Hanzu F, Mora M, Vicente A, Blanco Carrera C, De Miguel Novoa P, López García MDC, Manjón-Miguélez L, Rodríguez de Vera P, Del Castillo Tous M, Barahona San Millán R, Recasens M, Tomé Fernández-Ladreda M, Valdés N, Gracia Gimeno P, Robles Lazaro C, Michalopoulou T, Álvarez Escolá C, García Centeno R, Lamas C. Differences in intraoperative and surgical outcomes between normotensive pheochromocytomas and sympathetic paragangliomas (PPGLs) and hypertensive PPGLs: results from the PHEO-RISK STUDY. J Endocrinol Invest 2023; 46:805-814. [PMID: 36323983 DOI: 10.1007/s40618-022-01954-9] [Citation(s) in RCA: 5] [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: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE To compare the intraoperative and surgical outcomes of normotensive pheochromocytomas and sympathetic paragangliomas (PPGLs), hypertensive PPGLs and non-PPGL adrenal lesions. METHODS This a retrospective multicenter cohort study of patients with PPGLs from 18 tertiary hospitals. A control group of histologically confirmed adrenocortical adenomas (non-PPGL group) was selected to compare intraoperative and surgical outcomes with of the normotensive PPGLs. RESULTS Two hundred and ninety-six surgeries performed in 289 patients with PPGLs were included. Before surgery, 209 patients were classified as hypertensive PPGLs (70.6%) and 87 as normotensive PPGLs. A higher proportion of normotensive PPGLs than hypertensive PPGLs did not receive alpha presurgical blockade (P = 0.009). When we only considered those patients who received presurgical alpha blockers (200 hypertensive PPGLs and 76 normotensive PPGLs), hypertensive PPGLs had a threefold higher risk of intraoperative hypertensive crisis (OR 3.0 [95% 1.3-7.0]) and of hypotensive episodes (OR 2.9 [95% CI 1.2-6.7]) than normotensive PPGLs. When we compared normotensive PPGLs (n = 76) and non-PPGLs (n = 58), normotensive PPGLs had a fivefold higher risk of intraoperative complications (OR 5.3 [95% CI 1.9-14.9]) and a six times higher risk of postoperative complications (OR 6.1 [95% CI 1.7-21.6]) than non-PPGLs. CONCLUSION Although the risk of intraoperative hypertensive and hypotensive episodes in normotensive PPGLs is significantly lower than in hypertensive PPGLs, normotensive PPGLs have a greater risk of intraoperative and postoperative complications than non-PPGL adrenal lesions. Therefore, it is recommended to follow the standard of care for presurgical and anesthetic management of PPGLs also in normotensive PPGLs.
Collapse
Affiliation(s)
- M Araujo-Castro
- Endocrinology and Nutrition Department, Hospital Universitario Ramón y Cajal, Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Colmenar Viejo Street Km 9, 28034, Madrid, Spain.
- University of Alcalá, Madrid, Spain.
| | - I García Sanz
- General and Digestive Surgery Department, Hospital Universitario de La Princesa, Madrid, Spain
| | - C Mínguez Ojeda
- Urology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - M Calatayud
- Endocrinology and Nutrition Department, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - F Hanzu
- Endocrinology and Nutrition Department, Hospital Clinic, Barcelona, Spain
| | - M Mora
- Endocrinology and Nutrition Department, Hospital Clinic, Barcelona, Spain
| | - A Vicente
- Endocrinology and Nutrition Department, Hospital Universitario de Toledo, Toledo, Spain
| | - C Blanco Carrera
- Endocrinology and Nutrition Department, Hospital Universitario Príncipe de Asturias, Madrid, Spain
| | - P De Miguel Novoa
- Endocrinology and Nutrition Department, Hospital Clínico San Carlos, Madrid, Spain
| | | | - L Manjón-Miguélez
- Endocrinology and Nutrition Department, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - P Rodríguez de Vera
- Endocrinology and Nutrition Department, Hospital Universitario Virgen de La Macarena, Seville, Spain
| | - M Del Castillo Tous
- Endocrinology and Nutrition Department, Hospital Universitario Virgen de La Macarena, Seville, Spain
| | - R Barahona San Millán
- Endocrinology and Nutrition Department, Institut Català de la Salut Girona, Girona, Spain
| | - M Recasens
- Endocrinology and Nutrition Department, Institut Català de la Salut Girona, Girona, Spain
| | | | - N Valdés
- Endocrinology and Nutrition Department, Hospital Universitario de Cabueñes, Asturias, Spain
| | - P Gracia Gimeno
- Endocrinology and Nutrition Department, Hospital Royo Villanova, Zaragoza, Spain
| | - C Robles Lazaro
- Endocrinology and Nutrition Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - T Michalopoulou
- Department of Endocrinology and Nutrition, Joan XXIII University Hospital, Tarragona, Spain
| | - C Álvarez Escolá
- Endocrinology and Nutrition Department, Hospital Universitario La Paz, Madrid, Spain
| | - R García Centeno
- Endocrinology and Nutrition Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - C Lamas
- Endocrinology and Nutrition Department, Hospital Universitario de Albacete, Albacete, Spain
| |
Collapse
|
12
|
Gupta S, Erickson LA. Back to Biochemistry: Evaluation for and Prognostic Significance of SDH Mutations in Paragangliomas and Pheochromocytomas. Surg Pathol Clin 2023; 16:119-129. [PMID: 36739159 DOI: 10.1016/j.path.2022.09.011] [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] [Indexed: 02/05/2023]
Abstract
There is increasing recognition of the high prevalence of hereditary predisposition syndromes in patients diagnosed with paraganglioma/pheochromocytoma. It is widely acknowledged that germline pathogenic alterations of the succinate dehydrogenase complex genes (SDHA, SDHB, SDHC, SDHD, SDHAF2) contribute to the pathogenesis of most of these tumors. Herein, we have provided an update on the biology and diagnosis of succinate dehydrogenase-deficient paraganglioma/pheochromocytoma, including the molecular biology of the succinate dehydrogenase complex, mechanisms and consequences of inactivation of this complex, the prevalence of pathogenic alterations, and patterns of inheritance.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lori A Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Baechle JJ, Smith PM, Ortega CA, Wang TS, Solórzano CC, Kiernan CM. Clinical Predictors of Pseudohypoxia-Type Pheochromocytomas. Ann Surg Oncol 2022; 29:3536-3546. [PMID: 35233740 DOI: 10.1245/s10434-022-11419-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/16/2022] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Pheochromocytomas (PCCs) are rare tumors of neural crest origin with divergent transcriptional and metabolic profiles associated with mutational cluster types. Pseudohypoxia-type (PHT) PCCs have a poor prognosis; however diagnostic genetic testing is not always available. We aimed to investigate clinical parameters predictive of PHT PCCs. METHODS Patients who underwent resection and genetic testing for PCC at two academic centers from 2006-2020 were retrospectively studied. Patients with PHT mutations (SDH-AF2/B/C/D, VHL) were compared to non-pseudohypoxia-type (nonPHT) PCCs to identify widely available clinical parameters predictive of PHT PCCs. Demographic, clinical, and pathologic characteristics were compared using student's T and ANOVA tests. Operative hemodynamic instability was defined as systolic blood pressure (SBP) > 200 mmHg, SBP increase of > 30% relative to baseline, and/or heart rate (HR) > 110 bpm. Mann-Whitney U test was used to assess area under the curve (AUC), sensitivity, and specificity. Recursive partitioning was used to model predictive thresholds for PHT PCC and develop a predictive score. RESULTS Of the 79 patients included in the cohort, 17 (22%) had PHT and 62 (78%) had nonPHT PCCs. PCC patients with > 2 of the examined predictive clinical parameters (preoperative weight loss [> 10% body weight], elevated preoperative hematocrit [> 50%], normal baseline heart rate [< 100 bpm], and normal plasma metanephrines [< 0.60 nmol/L]) were more likely to have PHT PCCs (AUC = 0.831, sensitivity = 0.882, specificity = 0.694, all p < 0.001). CONCLUSIONS Widely available preoperative clinical parameters including indicators of erythropoiesis (hemoglobin, hematocrit, and red blood cell count), baseline heart rate, plasma metanephrines, and weight loss may be useful predictors of PHT PCCs and may help guide management of PCCs when genetic testing is unavailable/delayed.
Collapse
Affiliation(s)
- J J Baechle
- School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - P Marincola Smith
- Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C A Ortega
- School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - T S Wang
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - C C Solórzano
- Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C M Kiernan
- Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
14
|
Prinzi N, Corti F, Torchio M, Niger M, Antista M, Pagani F, Beninato T, Pulice I, Rossi RE, Coppa J, Cascella T, Giacomelli L, Di Bartolomeo M, Milione M, de Braud F, Pusceddu S. Metastatic pheochromocytomas and paragangliomas: where are we? TUMORI JOURNAL 2022; 108:526-540. [PMID: 35593402 DOI: 10.1177/03008916221078621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) can metastasize in approximately 15-20% of cases. This review discusses the available evidence on the biology and treatment of metastatic PPGLs. Chemotherapy is the first-line treatment option for this evolving and symptomatic disease. In patients with high MIBG uptake and positive PETGa-68, radiometabolic treatment may be considered. The efficacy of sunitinib has been shown in observational studies, and pembrolizumab has been evaluated in phase II clinical studies, while other agents investigated in this setting are anti-angiogenic drugs cabozantinib, dovitinib, axitinib and lenvatinib. As these agents' efficacy and safety data, alone or in combination, are scant and based on few treated patients, enrollment in clinical trials is mandatory. Future therapeutic options may be represented by DNA repair system inhibitors (such as olaparib), HIF2 inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Martina Torchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Teresa Beninato
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Iolanda Pulice
- Clinical Trial Center, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberta Elisa Rossi
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Pathophysiology and Organ Transplant, Università degli Studi di Milano, Milan, Italy
| | - Jorgelina Coppa
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso Cascella
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Massimo Milione
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy.,Oncology and Hemato-Oncology Department, Università degli Studi di Milano, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| |
Collapse
|
15
|
Martinelli S, Riverso M, Mello T, Amore F, Parri M, Simeone I, Mannelli M, Maggi M, Rapizzi E. SDHB and SDHD silenced pheochromocytoma spheroids respond differently to tumour microenvironment and their aggressiveness is inhibited by impairing stroma metabolism. Mol Cell Endocrinol 2022; 547:111594. [PMID: 35149119 DOI: 10.1016/j.mce.2022.111594] [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: 04/30/2021] [Revised: 01/11/2022] [Accepted: 02/04/2022] [Indexed: 12/18/2022]
Abstract
Germline mutations in more than 20 genes, including those encoding for the succinate dehydrogenase (SDH), predispose to rare tumours, such as pheochromocytoma/paraganglioma (PPGL). Despite encoding for the same enzymatic complex, SDHC and SDHD mutated PHEO/PGLs are generally benign, while up to 80% of SDHB mutated ones are malignant. In this study, we evaluated the different effects of tumour microenvironment on tumour cell migration/invasion, by co-culturing SDHB or SDHD silenced tumour spheroids with primary cancer-associated fibroblasts (CAFs). We observed that SDHD silenced spheroids had an intermediate migration pattern, compared to the highest migration capability of SDHB and the lowest one of the wild type (Wt) spheroids. Interestingly, we noticed that co-culturing Wt, SDHB and SDHD silenced spheroids with CAFs in low glucose (1 g/l) medium, caused a decreased migration of all the spheroids, but only for SDHB silenced ones this reduction was significant. Moreover, the collective migration, observed in high glucose (4.5 g/l) and characteristic of the SDHB silenced cells, was completely lost in low glucose. Importantly, migration could not be recovered even adding glucose (3.5 g/l) to low glucose conditioned medium. When we investigated cell metabolism, we found that low glucose concentration led to a reduction of oxygen consumption rate (OCR), basal and maximal oxidative metabolism, and ATP production only in CAFs, but not in tumour cells. These results suggest that CAFs metabolism impairment was responsible for the decreased invasion process of tumour cells, most likely preventing the release of the pro-migratory factors produced by CAFs. In conclusion, the interplay between CAFs and tumour cells is distinctive depending on the gene involved, and highlights the possibility to inhibit CAF-induced migration by impairing CAFs metabolism, indicating new potential therapeutic scenarios for medical therapy.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Maria Riverso
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Tommaso Mello
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Francesca Amore
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Matteo Parri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Irene Simeone
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine, University of Florence, Italy.
| |
Collapse
|
16
|
Nölting S, Bechmann N, Taieb D, Beuschlein F, Fassnacht M, Kroiss M, Eisenhofer G, Grossman A, Pacak K. Personalized Management of Pheochromocytoma and Paraganglioma. Endocr Rev 2022; 43:199-239. [PMID: 34147030 PMCID: PMC8905338 DOI: 10.1210/endrev/bnab019] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Pheochromocytomas/paragangliomas are characterized by a unique molecular landscape that allows their assignment to clusters based on underlying genetic alterations. With around 30% to 35% of Caucasian patients (a lower percentage in the Chinese population) showing germline mutations in susceptibility genes, pheochromocytomas/paragangliomas have the highest rate of heritability among all tumors. A further 35% to 40% of Caucasian patients (a higher percentage in the Chinese population) are affected by somatic driver mutations. Thus, around 70% of all patients with pheochromocytoma/paraganglioma can be assigned to 1 of 3 main molecular clusters with different phenotypes and clinical behavior. Krebs cycle/VHL/EPAS1-related cluster 1 tumors tend to a noradrenergic biochemical phenotype and require very close follow-up due to the risk of metastasis and recurrence. In contrast, kinase signaling-related cluster 2 tumors are characterized by an adrenergic phenotype and episodic symptoms, with generally a less aggressive course. The clinical correlates of patients with Wnt signaling-related cluster 3 tumors are currently poorly described, but aggressive behavior seems likely. In this review, we explore and explain why cluster-specific (personalized) management of pheochromocytoma/paraganglioma is essential to ascertain clinical behavior and prognosis, guide individual diagnostic procedures (biochemical interpretation, choice of the most sensitive imaging modalities), and provide personalized management and follow-up. Although cluster-specific therapy of inoperable/metastatic disease has not yet entered routine clinical practice, we suggest that informed personalized genetic-driven treatment should be implemented as a logical next step. This review amalgamates published guidelines and expert views within each cluster for a coherent individualized patient management plan.
Collapse
Affiliation(s)
- Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), CH-8091 Zurich, Switzerland.,Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, 13273 Marseille, France
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), CH-8091 Zurich, Switzerland.,Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany.,Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX2 6HG, UK.,Centre for Endocrinology, Barts and the London School of Medicine, London EC1M 6BQ, UK.,ENETS Centre of Excellence, Royal Free Hospital, London NW3 2QG, UK
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD 20847, USA
| |
Collapse
|
17
|
White G, Velusamy A, Anandappa S, Masucci M, Breen LA, Joshi M, McGowan B, Hubbard JGH, Obholzer R, Christodoulou D, Jacques A, Touska P, Hassan FU, Izatt L, Carroll PV. Tumour detection and outcomes of surveillance screening in SDHB and SDHD pathogenic variant carriers. Endocr Connect 2022; 11:EC-21-0602.R1. [PMID: 35060925 PMCID: PMC8859962 DOI: 10.1530/ec-21-0602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Succinate dehydrogenase subunit (SDHx) pathogenic variants predispose to phaeochromocytoma and paraganglioma (PPGL). Lifelong surveillance is recommended for all patients to enable prompt detection and treatment. There is currently limited evidence for optimal surveillance strategies in hereditary PPGL. We aim to detail the clinical presentation of PPGL in our cohort of non-index SDHB and SDHD pathogenic variant carriers. METHODS Retrospective analysis of medical and genetic records from a single tertiary referral centre identified SDHB or SDHD pathogenic variants in 74 non-index cases (56 SDHB and 18 SDHD). Surveillance screening for asymptomatic relatives consisted of annual plasma metanephrine measurement and whole-body MRI with contrast at 3-5 yearly intervals. RESULTS Twenty-three out of 74 non-index patients (10 SDHB and 13 SDHD) were diagnosed with PPGL, 17 patients through surveillance screening (24 tumours in total) and 6 diagnosed prior to commencement of cascade screening with symptomatic presentation. MRI with contrast identified PPGL in 22/24 screen-detected tumours and 5/24 tumours had elevated plasma metanephrine levels. Penetrance in non-index family members was 15.2 and 47.2% for SDHB carriers and 71.6 and 78.7% for SDHD carriers at age of 50 and 70 years, respectively. CONCLUSION Surveillance screening with combined biochemical testing and imaging enables early detection of PPGL in asymptomatic relatives with SDHx pathogenic variants. The presence of disease at first screen was significant in our cohort and hence further multi-centre long-term data are needed to inform counselling of family members undergoing lifelong surveillance.
Collapse
Affiliation(s)
- Gemma White
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Anand Velusamy
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Correspondence should be addressed to A Velusamy:
| | - Samantha Anandappa
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Michael Masucci
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Louise A Breen
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Mamta Joshi
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Barbara McGowan
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Johnathan G H Hubbard
- Department of Endocrine Surgery, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Rupert Obholzer
- Department of Ear, Nose and Throat Surgery, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | | | - Audrey Jacques
- Department of Radiology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Philip Touska
- Department of Radiology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Fahim-Ul Hassan
- Department of Nuclear Medicine, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Louise Izatt
- Department of Clinical Genetics, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Paul V Carroll
- Department of Diabetes and Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| |
Collapse
|
18
|
Constantinescu G, Preda C, Constantinescu V, Siepmann T, Bornstein SR, Lenders JWM, Eisenhofer G, Pamporaki C. Silent pheochromocytoma and paraganglioma: Systematic review and proposed definitions for standardized terminology. Front Endocrinol (Lausanne) 2022; 13:1021420. [PMID: 36325453 PMCID: PMC9618947 DOI: 10.3389/fendo.2022.1021420] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors with heterogeneous clinical presentations and potential lethal outcomes. The diagnosis is based on clinical suspicion, biochemical testing, imaging and histopathological confirmation. Increasingly widespread use of imaging studies and surveillance of patients at risk of PPGL due to a hereditary background or a previous tumor is leading to the diagnosis of these tumors at an early stage. This has resulted in an increasing use of the term "silent" PPGL. This term and other variants are now commonly found in the literature without any clear or unified definition. Among the various terms, "clinically silent" is often used to describe the lack of signs and symptoms associated with catecholamine excess. Confusion arises when these and other terms are used to define the tumors according to their ability to synthesize and/or release catecholamines in relation to biochemical test results. In such cases the term "silent" and other variants are often inappropriately and misleadingly used. In the present analysis we provide an overview of the literature and propose standardized terminology in an attempt at harmonization to facilitate scientific communication.
Collapse
Affiliation(s)
- Georgiana Constantinescu
- Department of Endocrinology and Diabetes, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Endocrinology, Grigore T. Popa University, Iasi, Romania
- Department of Health Care Sciences, Center for Clinical Research and Management Education, Dresden Inter-national University, Dresden, Germany
- *Correspondence: Christina Pamporaki, ; Georgiana Constantinescu,
| | - Cristina Preda
- Department of Endocrinology, Grigore T. Popa University, Iasi, Romania
| | - Victor Constantinescu
- Center of Clinical Neuroscience, University Clinic Carl-Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Timo Siepmann
- Department of Health Care Sciences, Center for Clinical Research and Management Education, Dresden Inter-national University, Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Endocrinology and Diabetes, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Health Care Sciences, Center for Clinical Research and Management Education, Dresden International University, Dresden, Germany
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Jacques W. M. Lenders
- Department of Endocrinology and Diabetes, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Graeme Eisenhofer
- Department of Endocrinology and Diabetes, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University of Dresden, Dresden, Germany
| | - Christina Pamporaki
- Department of Endocrinology and Diabetes, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Christina Pamporaki, ; Georgiana Constantinescu,
| |
Collapse
|
19
|
Hiromasa T, Wakabayashi H, Kayano D, Inaki A, Watanabe S, Mori H, Akatani N, Yamase T, Kunita Y, Saito S, Kinuya S. Prognostic factors for refractory pheochromocytoma and paraganglioma after 131I-metaiodobenzylguanidine therapy. Ann Nucl Med 2022; 36:61-69. [PMID: 34647244 DOI: 10.1007/s12149-021-01685-6] [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: 08/11/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Given the rarity of refractory pheochromocytoma and paraganglioma (PPGL), outcomes and prognostic factors after 131I-metaiodobenzylguanidine (131I-mIBG) treatment still remain unclear. Therefore, this study evaluated whether baseline characteristics at initial 131I-mIBG therapy and imaging response to repeated 131I-mIBG therapy could be prognostic factors for refractory PPGL. METHODS All patients [n = 59 (male/female = 35/24), median age; 49.3 years] with refractory PPGL who received 131I-mIBG therapy at our institution between September 2009 and September 2019 were retrospectively reviewed for the effects of the following factors on overall survival: age, sex, hypertension, diabetes mellitus, palpitations, constipation, cancer pain, catecholamines values, past history of therapy (external beam radiation for bone metastasis, operation, and chemotherapy), metastasis sites, and response to 131I-mIBG treatments. RESULTS Throughout the follow-up period, 18 patients died from disease exacerbation. The estimated 5- and 10-year survival rates were 79.4% and 67.2% from the initial diagnoses of refractory PPGL and 68.5% and 49.9% from the first 131I-mIBG therapy, respectively. The multivariate Cox proportional hazards model showed that progressive disease (PD) [hazard ratio (HR) 96.3, P = 0.011] and constipation (HR 8.2, P = 0.024) were adverse prognostic factors for overall survival after initial 131I-mIBG therapy. The log-rank test demonstrated that PD in response to 131I-mIBG therapies (P < 0.0001) and constipation (P < 0.01) were correlated with poor survival rates. CONCLUSIONS Response to repeated 131I-mIBG treatment can be a strong predictor of prognosis after initial 131I-mIBG therapy for refractory PPGL. Repeated 131I-mIBG therapy may be a good option for controlling refractory PPGL.
Collapse
Affiliation(s)
- Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Anri Inaki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Satoru Watanabe
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hiroshi Mori
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Norihito Akatani
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Takafumi Yamase
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Yuji Kunita
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Shintaro Saito
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
20
|
Takács-Vellai K, Farkas Z, Ősz F, Stewart GW. Model systems in SDHx-related pheochromocytoma/paraganglioma. Cancer Metastasis Rev 2021; 40:1177-1201. [PMID: 34957538 PMCID: PMC8825606 DOI: 10.1007/s10555-021-10009-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/04/2021] [Indexed: 11/17/2022]
Abstract
Pheochromocytoma (PHEO) and paraganglioma (PGL) (together PPGL) are tumors with poor outcomes that arise from neuroendocrine cells in the adrenal gland, and sympathetic and parasympathetic ganglia outside the adrenal gland, respectively. Many follow germline mutations in genes coding for subunits of succinate dehydrogenase (SDH), a tetrameric enzyme in the tricarboxylic acid (TCA) cycle that both converts succinate to fumarate and participates in electron transport. Germline SDH subunit B (SDHB) mutations have a high metastatic potential. Herein, we review the spectrum of model organisms that have contributed hugely to our understanding of SDH dysfunction. In Saccharomyces cerevisiae (yeast), succinate accumulation inhibits alpha-ketoglutarate-dependent dioxygenase enzymes leading to DNA demethylation. In the worm Caenorhabditis elegans, mutated SDH creates developmental abnormalities, metabolic rewiring, an energy deficit and oxygen hypersensitivity (the latter is also found in Drosophila melanogaster). In the zebrafish Danio rerio, sdhb mutants display a shorter lifespan with defective energy metabolism. Recently, SDHB-deficient pheochromocytoma has been cultivated in xenografts and has generated cell lines, which can be traced back to a heterozygous SDHB-deficient rat. We propose that a combination of such models can be efficiently and effectively used in both pathophysiological studies and drug-screening projects in order to find novel strategies in PPGL treatment.
Collapse
Affiliation(s)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Budapest, Hungary
| | - Fanni Ősz
- Department of Biological Anthropology, Eötvös Loránd University, Budapest, Hungary
| | - Gordon W Stewart
- Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
21
|
Snezhkina A, Pavlov V, Dmitriev A, Melnikova N, Kudryavtseva A. Potential Biomarkers of Metastasizing Paragangliomas and Pheochromocytomas. Life (Basel) 2021; 11:1179. [PMID: 34833055 PMCID: PMC8619623 DOI: 10.3390/life11111179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 02/07/2023] Open
Abstract
Paragangliomas and pheochromocytomas (PPGLs) are rare neuroendocrine tumors originating from paraganglionic tissue in many sites of the body. Most PPGLs are characterized by nonaggressive behavior but all of them have the potential to metastasize. PPGLs represent a great diagnostic dilemma as it is difficult to recognize tumors that are likely to be metastasizing; criteria of malignancy can be found both in benign and metastatic forms. This review aims to analyze the current knowledge of the nature of metastasizing PPGLs paying particular attention to head and neck paragangliomas (HNPGLs). Potential predictors of the malignancy risk for PPGLs were summarized and discussed. These data may also help in the development of diagnostic and prognostic strategies, as well as in the identification of novel potential therapeutic targets for patients with PPGLs.
Collapse
Affiliation(s)
- Anastasiya Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (V.P.); (A.D.); (N.M.)
| | | | | | | | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (V.P.); (A.D.); (N.M.)
| |
Collapse
|
22
|
Amar L, Pacak K, Steichen O, Akker SA, Aylwin SJB, Baudin E, Buffet A, Burnichon N, Clifton-Bligh RJ, Dahia PLM, Fassnacht M, Grossman AB, Herman P, Hicks RJ, Januszewicz A, Jimenez C, Kunst HPM, Lewis D, Mannelli M, Naruse M, Robledo M, Taïeb D, Taylor DR, Timmers HJLM, Treglia G, Tufton N, Young WF, Lenders JWM, Gimenez-Roqueplo AP, Lussey-Lepoutre C. International consensus on initial screening and follow-up of asymptomatic SDHx mutation carriers. Nat Rev Endocrinol 2021; 17:435-444. [PMID: 34021277 PMCID: PMC8205850 DOI: 10.1038/s41574-021-00492-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
Approximately 20% of patients diagnosed with a phaeochromocytoma or paraganglioma carry a germline mutation in one of the succinate dehydrogenase (SDHx) genes (SDHA, SDHB, SDHC and SDHD), which encode the four subunits of the SDH enzyme. When a pathogenic SDHx mutation is identified in an affected patient, genetic counselling is proposed for first-degree relatives. Optimal initial evaluation and follow-up of people who are asymptomatic but might carry SDHx mutations have not yet been agreed. Thus, we established an international consensus algorithm of clinical, biochemical and imaging screening at diagnosis and during surveillance for both adults and children. An international panel of 29 experts from 12 countries was assembled, and the Delphi method was used to reach a consensus on 41 statements. This Consensus Statement covers a range of topics, including age of first genetic testing, appropriate biochemical and imaging tests for initial tumour screening and follow-up, screening for rare SDHx-related tumours and management of elderly people who have an SDHx mutation. This Consensus Statement focuses on the management of asymptomatic SDHx mutation carriers and provides clinicians with much-needed guidance. The standardization of practice will enable prospective studies in the near future.
Collapse
Affiliation(s)
- Laurence Amar
- Paris University, Hypertension unit, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
- INSERM, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD, USA
| | - Olivier Steichen
- Sorbonne University, Department of Internal Medicine, Hôpital Tenon, AP-HP, Paris, France
| | - Scott A Akker
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | | | - Eric Baudin
- Gustave Roussy Institute and Paris Saclay University, Villejuif, France
| | - Alexandre Buffet
- INSERM, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Genetics Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Nelly Burnichon
- INSERM, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Genetics Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Roderick J Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Patricia L M Dahia
- Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Martin Fassnacht
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ashley B Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- NET Unit, Royal Free Hospital, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
| | - Philippe Herman
- ENT unit, Lariboisière Hospital, AP-HP, University of Paris, Paris, France
| | - Rodney J Hicks
- Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrzej Januszewicz
- Department of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Henricus P M Kunst
- Department of ENT, Radboud University Medical Center, Nijmegen, Netherlands
- Maastricht University Medical Center, Maastricht, Netherlands
| | - Dylan Lewis
- King's College Hospital NHS Foundation Trust, London, UK
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Mitsuhide Naruse
- Endocrine Center, Ijinkai Takeda General Hospital and Clinical Research Institute, NHO Kyoto Medical Center, Kyoto, Japan
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group. Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - David Taïeb
- Aix-Marseille University, La Timone university hospital, European Center for Research in Medical Imaging, Marseille, France
| | - David R Taylor
- King's College Hospital NHS Foundation Trust, London, UK
| | - Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Giorgio Treglia
- Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nicola Tufton
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - William F Young
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anne-Paule Gimenez-Roqueplo
- INSERM, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Genetics Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Charlotte Lussey-Lepoutre
- INSERM, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.
- Sorbonne University, Nuclear medicine department, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| |
Collapse
|
23
|
Hirose R, Tsurutani Y, Sugisawa C, Inoue K, Suematsu S, Nagata M, Hasegawa N, Kakuta Y, Yonamine M, Takekoshi K, Kimura N, Saito J, Nishikawa T. Hereditary pheochromocytoma/paraganglioma syndrome with a novel mutation in the succinate dehydrogenase subunit B gene in a Japanese family: two case reports. J Med Case Rep 2021; 15:282. [PMID: 34020699 PMCID: PMC8140422 DOI: 10.1186/s13256-021-02852-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pheochromocytoma and paraganglioma caused by succinate dehydrogenase gene mutations is called hereditary pheochromocytoma/paraganglioma syndrome. In particular, succinate dehydrogenase subunit B mutations are important because they are strongly associated with the malignant behavior of pheochromocytoma and paraganglioma . This is a case report of a family of hereditary pheochromocytoma/paraganglioma syndrome carrying a novel mutation in succinate dehydrogenase subunit B. CASE PRESENTATION A 19-year-old Japanese woman, whose father died of metastatic paraganglioma, was diagnosed with abdominal paraganglioma, and underwent total resection. Succinate dehydrogenase subunit B genetic testing detected a splice-site mutation, c.424-2delA, in her germline and paraganglioma tissue. Afterwards, the same succinate dehydrogenase subunit B mutation was detected in her father's paraganglioma tissues. In silico analysis predicted the mutation as "disease causing." She is under close follow-up, and no recurrence or metastasis has been observed for 4 years since surgery. CONCLUSIONS We detected a novel succinate dehydrogenase subunit B mutation, c.424-2delA, in a Japanese family afflicted with hereditary pheochromocytoma/paraganglioma syndrome and found the mutation to be responsible for hereditary pheochromocytoma/paraganglioma syndrome. This case emphasizes the importance of performing genetic testing for patients with pheochromocytoma and paraganglioma suspected of harboring the succinate dehydrogenase subunit B mutation (that is, metastatic, extra-adrenal, multiple, early onset, and family history of pheochromocytoma and paraganglioma) and offer surveillance screening to mutation carriers.
Collapse
Affiliation(s)
- Rei Hirose
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Yuya Tsurutani
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan.
| | - Chiho Sugisawa
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Kosuke Inoue
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan.,Department of Epidemiology, UCLA Fielding School of Public Health, 650 Charles E. Young Dr. South, 16-035 Center for Health Sciences, Los Angeles, CA, USA
| | - Sachiko Suematsu
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Maki Nagata
- Department of Urology, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Naoki Hasegawa
- Department of Pathology, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Yukio Kakuta
- Department of Pathology, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Masato Yonamine
- Laboratory of Laboratory/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Kazuhiro Takekoshi
- Laboratory of Laboratory/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Noriko Kimura
- Department of Diagnostic Pathology, National Hospital Organization Hakodate Hospital, 18-16 Kawahara-cho, Hakodate, Hokkaido, 041-8512, Japan
| | - Jun Saito
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| | - Tetsuo Nishikawa
- Endocrinology and Diabetes Center, Yokohama Rosai Hospital, 3211 Kozukue-cho, Kouhoku-ku, Yokohama, Kanagawa, 222-0036, Japan
| |
Collapse
|
24
|
Saskői É, Hujber Z, Nyírő G, Likó I, Mátyási B, Petővári G, Mészáros K, Kovács AL, Patthy L, Supekar S, Fan H, Sváb G, Tretter L, Sarkar A, Nazir A, Sebestyén A, Patócs A, Mehta A, Takács-Vellai K. The SDHB Arg230His mutation causing familial paraganglioma alters glycolysis in a new Caenorhabditis elegans model. Dis Model Mech 2020; 13:dmm044925. [PMID: 32859697 PMCID: PMC7578352 DOI: 10.1242/dmm.044925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
The conserved B-subunit of succinate dehydrogenase (SDH) participates in the tricarboxylic acid cycle (TCA) cycle and mitochondrial electron transport. The Arg230His mutation in SDHB causes heritable pheochromocytoma/paraganglioma (PPGL). In Caenorhabditiselegans, we generated an in vivo PPGL model (SDHB-1 Arg244His; equivalent to human Arg230His), which manifests delayed development, shortened lifespan, attenuated ATP production and reduced mitochondrial number. Although succinate is elevated in both missense and null sdhb-1(gk165) mutants, transcriptomic comparison suggests very different causal mechanisms that are supported by metabolic analysis, whereby only Arg244His (not null) worms demonstrate elevated lactate/pyruvate levels, pointing to a missense-induced, Warburg-like aberrant glycolysis. In silico predictions of the SDHA-B dimer structure demonstrate that Arg230His modifies the catalytic cleft despite the latter's remoteness from the mutation site. We hypothesize that the Arg230His SDHB mutation rewires metabolism, reminiscent of metabolic reprogramming in cancer. Our tractable model provides a novel tool to investigate the metastatic propensity of this familial cancer and our approach could illuminate wider SDH pathology.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Éva Saskői
- Department of Biological Anthropology, Eötvös Lorand University, Budapest H-1117, Hungary
| | - Zoltán Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest H-1085, Hungary
| | - Gábor Nyírő
- HAS-SE Momentum Hereditary Endocrine Tumour Syndromes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1089, Hungary
| | - István Likó
- HAS-SE Momentum Hereditary Endocrine Tumour Syndromes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1089, Hungary
| | - Barbara Mátyási
- Department of Biological Anthropology, Eötvös Lorand University, Budapest H-1117, Hungary
| | - Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest H-1085, Hungary
| | - Katalin Mészáros
- HAS-SE Momentum Hereditary Endocrine Tumour Syndromes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1089, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest H-1089, Hungary
| | - Attila L Kovács
- Department of Anatomy, Cell and Developmental Biology, Eötvös Lorand University, Budapest H-1117, Hungary
| | - László Patthy
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | - Shreyas Supekar
- Bioinformatics Institute, Agency for Science, Technology and Research, 138671 Singapore
| | - Hao Fan
- Bioinformatics Institute, Agency for Science, Technology and Research, 138671 Singapore
| | - Gergely Sváb
- Department of Medical Biochemistry, MTA-SE Laboratory for Neurobiochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - László Tretter
- Department of Medical Biochemistry, MTA-SE Laboratory for Neurobiochemistry, Semmelweis University, Budapest H-1094, Hungary
| | - Arunabh Sarkar
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Aamir Nazir
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest H-1085, Hungary
| | - Attila Patócs
- HAS-SE Momentum Hereditary Endocrine Tumour Syndromes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1089, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest H-1089, Hungary
| | - Anil Mehta
- Division of Medical Sciences, Ninewells Hospital Medical School, University of Dundee, Dundee DD1 1NH, UK
| | | |
Collapse
|
25
|
Kudryavtseva AV, Kalinin DV, Pavlov VS, Savvateeva MV, Fedorova MS, Pudova EA, Kobelyatskaya AA, Golovyuk AL, Guvatova ZG, Razmakhaev GS, Demidova TB, Simanovsky SA, Slavnova EN, Poloznikov AА, Polyakov AP, Melnikova NV, Dmitriev AA, Krasnov GS, Snezhkina AV. Mutation profiling in eight cases of vagal paragangliomas. BMC Med Genomics 2020; 13:115. [PMID: 32948195 PMCID: PMC7500026 DOI: 10.1186/s12920-020-00763-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/05/2020] [Indexed: 12/25/2022] Open
Abstract
Background Vagal paragangliomas (VPGLs) belong to a group of rare head and neck neuroendocrine tumors. VPGLs arise from the vagus nerve and are less common than carotid paragangliomas. Both diagnostics and therapy of the tumors raise significant challenges. Besides, the genetic and molecular mechanisms behind VPGL pathogenesis are poorly understood. Methods The collection of VPGLs obtained from 8 patients of Russian population was used in the study. Exome library preparation and high-throughput sequencing of VPGLs were performed using an Illumina technology. Results Based on exome analysis, we identified pathogenic/likely pathogenic variants of the SDHx genes, frequently mutated in paragangliomas/pheochromocytomas. SDHB variants were found in three patients, whereas SDHD was mutated in two cases. Moreover, likely pathogenic missense variants were also detected in SDHAF3 and SDHAF4 genes encoding for assembly factors for the succinate dehydrogenase (SDH) complex. In a patient, we found a novel variant of the IDH2 gene that was predicted as pathogenic by a series of algorithms used (such as SIFT, PolyPhen2, FATHMM, MutationTaster, and LRT). Additionally, pathogenic/likely pathogenic variants were determined for several genes, including novel genes and some genes previously reported as associated with different types of tumors. Conclusions Results indicate a high heterogeneity among VPGLs, however, it seems that driver events in most cases are associated with mutations in the SDHx genes and SDH assembly factor-coding genes that lead to disruptions in the SDH complex.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Dmitry V Kalinin
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladislav S Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria V Savvateeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Alexander L Golovyuk
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Zulfiya G Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Razmakhaev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Tatiana B Demidova
- A. N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Sergey A Simanovsky
- A. N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Elena N Slavnova
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey А Poloznikov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey P Polyakov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
26
|
Därr R, Kater J, Sekula P, Bausch B, Krauss T, Bode C, Walz G, Neumann HP, Zschiedrich S. Clinical decision making in small non-functioning VHL-related incidentalomas. Endocr Connect 2020; 9:834-844. [PMID: 32869749 PMCID: PMC7487196 DOI: 10.1530/ec-20-0208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 01/26/2023]
Abstract
The optimal treatment strategy for patients with small non-functioning VHL-related incidentalomas is unclear. We searched the Freiburg VHL registry for patients with radiologic evidence of pheochromocytoma/paraganglioma (PHEO/PGL). In total, 176 patients with single, multiple, and recurrent tumours were identified (1.84 tumours/patient, range 1-8). Mean age at diagnosis was 32 ± 16 years. Seventy-four percent of tumours were localised to the adrenals. Mean tumour diameter was 2.42 ± 2.27 cm, 46% were <1.5 cm. 24% of tumours were biochemically inactive. Inactive tumours were significantly smaller than active PHEO/PGL at diagnosis (4.16 ± 2.80 cm vs 1.43 ± 0.45 cm; P < 0.025) and before surgery (4.89 ± 3.47 cm vs 1.36 ± 0.43 cm; P < 0.02). Disease was stable in 67% of 21 patients with evaluable tumours ≤1.5 cm according to RECIST and progressed in 7. Time till surgery in these patients was 29.5 ± 20.0 months. A total of 155 patients underwent surgery. PHEO/PGL was histologically excluded in 4 and proven in 151. Of these, one had additional metastatic disease, one harboured another tumour of a different type, and in 2 a second surgery for suspected disease recurrence did not confirm PHEO/PGL. Logistic regression analysis revealed 50% probability for a positive/negative biochemical test result at 1.8 cm tumour diameter. Values of a novel symptom score were positively correlated with tumour size (Rs = 0.46, P < 0.0001) and together with a positive biochemistry a linear size predictor (P < 0.01). Results support standardised clinical assessment and measurement of tumour size and metanephrines in VHL patients with non-functioning incidentalomas <1.5 cm at one year following diagnosis and at individualised intervals thereafter depending on evolving growth dynamics, secretory activity and symptomatology.
Collapse
Affiliation(s)
- Roland Därr
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Correspondence should be addressed to R Därr:
| | - Jonas Kater
- Department of Medicine IV, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, University Medical Center Freiburg, Freiburg, Germany
| | - Birke Bausch
- Department of Medicine II, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Krauss
- Department of Radiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut P Neumann
- Section for Preventive Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Zschiedrich
- Department of Medicine IV, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
27
|
Main AM, Rossing M, Borgwardt L, Grønkær Toft B, Rasmussen ÅK, Feldt-Rasmussen U. Genotype-phenotype associations in PPGLs in 59 patients with variants in SDHX genes. Endocr Connect 2020; 9:793-803. [PMID: 32688340 PMCID: PMC7487185 DOI: 10.1530/ec-20-0279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/19/2020] [Indexed: 12/31/2022]
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are tumours of the adrenal medulla and extra-adrenal sympathetic nervous system which often secrete catecholamines. Variants of the SDHX (SDHA, -AF2, -B, -C, -D) genes are a frequent cause of familial PPGLs. In this study from a single tertiary centre, we aimed to characterise the genotype-phenotype associations in patients diagnosed with germline variants in SDHX genes. We also assessed whether systematic screening of family members resulted in earlier detection of tumours. The study cohort comprised all individuals (n = 59) diagnosed with a rare variant in SDHX during a 13-year period. Patient- and pathology records were checked for clinical characteristics and histopathological findings. We found distinct differences in the clinical and histopathological characteristics between genetic variants in SDHB. We identified two SDHB variants with distinct phenotypical patterns. Family screening for SDHB variants resulted in earlier detection of tumours in two families. Patients with SDHA, SDHC and SDHD variants also had malignant phenotypes, underlining the necessity for a broad genetic screening of the proband. Our study corroborates previous findings of poor prognostic markers and found that the genetic variants and clinical phenotype are linked and, therefore, useful in the decision of clinical follow-up. Regular tumour screening of carriers of pathogenic variants may lead to an earlier diagnosis and expected better prognosis. The development of a combined algorithm with clinical, genetic, morphological, and biochemical factors may be the future for improved clinical risk stratification, forming a basis for larger multi-centre follow up studies.
Collapse
Affiliation(s)
- Ailsa Maria Main
- Department of Medical Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Correspondence should be addressed to A M Main or U Feldt-Rasmussen: or
| | - Maria Rossing
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Line Borgwardt
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Birgitte Grønkær Toft
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Åse Krogh Rasmussen
- Department of Medical Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, and Faculty of Health, Institute of Clinical and Scientific Research, Copenhagen, Denmark
- Correspondence should be addressed to A M Main or U Feldt-Rasmussen: or
| |
Collapse
|
28
|
Chen H, Yao W, He Q, Yu X, Bian B. Identification of a novel SDHB c.563 T > C mutation responsible for Paraganglioma syndrome and genetic analysis of the SDHB gene in China: a case report. BMC MEDICAL GENETICS 2020; 21:116. [PMID: 32460727 PMCID: PMC7254674 DOI: 10.1186/s12881-020-01049-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/11/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Pheochromocytoma/paraganglioma (PPGL) is a rare neuroendocrine tumor. Succinate dehydrogenase (SDH) deficiency has been confirmed to be associated with PPGL in various studies. SDHB mutations play an important role in PPGL. However, genetic screening of PPGL patients has not been widely carried out in clinics in China, and only a few related studies have been reported. CASE PRESENTATION We report a case of a 23-year-old woman with paraganglioma (PGL) caused by a novel missense SDHB mutation, c.563 T > C (p.Leu188Pro), who presented with paroxysmal hypertension. Computed tomography (CT) and magnetic resonance imaging (MRI) revealed a PGL in the right retroperitoneum and no metastasis. The patient was treated with surgical excision and did not have postsurgerical paroxysmal hypertension. In addition, we searched the literature related to variations in SDHB genes in Chinese patients with PPGL using multiple online databases, including PubMed, China Hospital Knowledge Database and Wanfang Data. Ultimately, 14 studies (published between 2006 and 2019) comprising 34 cases of SDHB-related PGL or pheochromocytoma (PCC) were found. In total, 35 patients were enrolled in this study, and 25 mutations were identified. The common genetic alterations of SDHB in China were c.136C > T (11.4%), c.18C > A (11.4%) and c.725G > A (8.5%). Some carriers of SDHB mutations (28.1%) developed metastatic PPGL, and a high frequency of head and neck PGLs (HNPGLs) (59.4%) was reported. CONCLUSIONS We describe a classic case with a novel SDHB c.563 T > C mutation. Based on our literature review, common SDHB gene mutations in Chinese PPGL patients are c.136C > T, c.18C > A and c.725G > A.
Collapse
Affiliation(s)
- Heye Chen
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Wei Yao
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Xuefang Yu
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, 300070, China
| | - Bo Bian
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, 300070, China.
| |
Collapse
|
29
|
Abstract
Introduction Paragangliomas are rare endocrine tumors that arise from the extra-adrenal autonomic paraganglia and sympathetic paragangliomas usually secret catecholamines and are located in the sympathetic paravertebral ganglia of thorax, abdomen, and pelvis. In contrast, most parasympathetic paragangliomas are nonfunctional and located along the glossopharyngeal and vagal nerves in the neck and at the base of the skull. Such neoplasms, although rare, are clinically important because they may recur after surgical resection and 10% of them give rise to metastases causing death with the lymphatic nodes, bones, liver, and lungs being the most common locations. Case presentation We present a case of a 26-year-old male patient that was diagnosed with paraganglioma of the right-frontal lobe infiltrating the falx and frontal bone which was diagnosed after suffering from a headache and abnormal vision. On initial work-up he was found to have right pulmonary nodules that increased in size after follow up and other nodules appeared in the contralateral lung. He underwent subtotal resection of the brain tumor and complete resection of the bilateral pulmonary nodules. Conclusion To our knowledge, paraganglioma is considered to be a rare entity in the central nervous system with very few cases being reported in the supratentorial region and no cases were reported of metastatic such paraganglioma to the lung.
Collapse
|
30
|
Withey SJ, Perrio S, Christodoulou D, Izatt L, Carroll P, Velusamy A, Obholzer R, Lewington V, Jacques AET. Imaging Features of Succinate Dehydrogenase-deficient Pheochromocytoma-Paraganglioma Syndromes. Radiographics 2020; 39:1393-1410. [PMID: 31498738 DOI: 10.1148/rg.2019180151] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pheochromocytoma (PC) and paraganglioma (PGL) are rare neuroendocrine tumors that occur throughout the body from the base of the skull to the pelvis. Sympathetic catecholamine-secreting tumors may be associated with hyperadrenergic symptoms and long-term morbidity if they are untreated. Typically biochemically silent, head and neck PGLs may result in cranial nerve palsies and symptoms due to localized mass effect. Tumors can arise sporadically or as part of an inheritable PC-PGL syndrome. Up to 40% of tumors are recognized to be associated with germline mutations in an increasing array of susceptibility genes, including those that appear to arise sporadically. Most commonly, up to 25% of all PC-PGLs are associated with mutations in one of the succinate dehydrogenase (SDH) enzyme subunit genes. The resulting familial PC-PGL syndrome varies according to the affected enzyme subunit (most commonly SDHB and SDHD mutations) with respect to tumor prevalence, location, age of onset, and risk of malignancy. Patients with SDH enzyme mutations have increased lifetime risk of developing multifocal tumors and malignancy. Early recognition of individuals at high risk, genetic testing, screening of family members, and lifelong surveillance programs are recommended, but not without health, economic, and psychologic implications. Anatomic and functional imaging is key to diagnosis, staging, treatment planning, and lifelong surveillance of these individuals. Radiologists must be aware of the imaging appearance of these varied tumors.©RSNA, 2019.
Collapse
Affiliation(s)
- Samuel Joseph Withey
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Stephen Perrio
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Dimitra Christodoulou
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Louise Izatt
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Paul Carroll
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Anand Velusamy
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Rupert Obholzer
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Valerie Lewington
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| | - Audrey Eleanor Therese Jacques
- From the Departments of Radiology (S.J.W., S.P., D.C., A.E.T.J.), Genetics (L.I.), Endocrinology (P.C., A.V.), Ear, Nose, and Throat Surgery (R.O.), and Nuclear Medicine (V.L.), Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, Level 1, Lambeth Wing, London SE1 7EH, England; and Division of Imaging Sciences and Biomedical Engineering, King's College London, London, England (V.L.)
| |
Collapse
|
31
|
Jochmanova I, Abcede AMT, Guerrero RJS, Malong CLP, Wesley R, Huynh T, Gonzales MK, Wolf KI, Jha A, Knue M, Prodanov T, Nilubol N, Mercado-Asis LB, Stratakis CA, Pacak K. Clinical characteristics and outcomes of SDHB-related pheochromocytoma and paraganglioma in children and adolescents. J Cancer Res Clin Oncol 2020; 146:1051-1063. [PMID: 32062700 DOI: 10.1007/s00432-020-03138-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 01/12/2023]
Abstract
PURPOSE Pheochromocytomas/paragangliomas (PHEOs/PGLs) are rare in children with only a few SDHB mutation-related cases. Previous studies on children were conducted in small cohorts. This large set of pediatric patients provides robust data in the evaluation of clinical outcomes. METHODS Sixty-four pediatric PHEO/PGL patients with SDHB germline mutations were included in the present study. The clinical presentation, disease course, and survival rate were evaluated. RESULTS Thirty-eight males and 26 females were diagnosed with PHEO/PGL at a median age of 13 years. The majority of patients displayed norepinephrine hypersecretion and 73.44% initially presented with a solitary tumor. Metastases developed in 70% of patients at the median age of 16 years and were mostly diagnosed first 2 years and in years 12-18 post-diagnosis. The presence of metastases at the time of diagnosis had a strong negative impact on survival in males but not in females. The estimated 5-, 10-, and 20-year survival rates were 100%, 97.14%, and 77.71%, respectively. CONCLUSION The present report has highlighted several important aspects in the management of pediatric patients with SDHB mutations associated-PHEO/PGL. Initial diagnostic evaluation of SDHB mutation carriers should be started at age of 5-6 years with initial work-up focusing on abdominal region. Thorough follow-up is crucial first 2 years post-diagnosis and more frequent follow-ups are needed in years 10-20 post-diagnosis due to the increased risk of metastases. Although this age group developed metastasis as early as 5 years from diagnosis, we have shown that the overall 20-year prognosis and survival are good.
Collapse
Affiliation(s)
- Ivana Jochmanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- 1st Department of Internal Medicine, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - April Melody T Abcede
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Section of Endocrinology and Metabolism, Department of Medicine, University of Santo Tomas Hospital, 1008, Manila, Philippines
| | - Ruby Jane S Guerrero
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Section of Endocrinology and Metabolism, Department of Medicine, University of Santo Tomas Hospital, 1008, Manila, Philippines
| | - Chandy Lou P Malong
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
- Section of Endocrinology and Metabolism, Department of Medicine, University of Santo Tomas Hospital, 1008, Manila, Philippines
| | - Robert Wesley
- Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thanh Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Melissa K Gonzales
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Katherine I Wolf
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marianne Knue
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tamara Prodanov
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Leilani B Mercado-Asis
- Section of Endocrinology and Metabolism, Department of Medicine, University of Santo Tomas Hospital, 1008, Manila, Philippines
| | - Constantine A Stratakis
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Eunice Kennedy Shriver NICHD, NIH, Building 10, CRC, Room 1E-3140, 10 Center Drive MSC-1109, Bethesda, MD, 20892-1109, USA.
| |
Collapse
|
32
|
Neppala P, Banerjee S, Fanta PT, Yerba M, Porras KA, Burgoyne AM, Sicklick JK. Current management of succinate dehydrogenase-deficient gastrointestinal stromal tumors. Cancer Metastasis Rev 2020; 38:525-535. [PMID: 31773431 DOI: 10.1007/s10555-019-09818-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are increasingly recognized as having diverse biology. With the development of tyrosine kinase inhibitors molecularly matched to oncogenic KIT and PDGFRA mutations, GISTs have become a quintessential model for precision oncology. However, about 5-10% of GIST lack these driver mutations and are deficient in succinate dehydrogenase (SDH), an enzyme that converts succinate to fumarate. SDH deficiency leads to accumulation of succinate, an oncometabolite that promotes tumorigenesis. SDH-deficient GISTs are clinically unique in that they generally affect younger patients and are associated with GIST-paraganglioma hereditary syndrome, also known as Carney-Stratakis Syndrome. SDH-deficient GISTs are generally resistant to tyrosine-kinase inhibitors, the standard treatment for advanced or metastatic GIST. Thus, surgical resection is the mainstay of treatment for localized disease, but recurrence is common. Clinical trials are currently underway investigating systemic agents for treatment of advanced SDH-deficient GIST. However, further studies are warranted to improve our understanding of SDH-deficient GIST disease biology, natural history, surgical approaches, and novel therapeutics.
Collapse
Affiliation(s)
- Pushpa Neppala
- UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sudeep Banerjee
- Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Paul T Fanta
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Division of Hematology-Oncology, Department of Medicine, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Mayra Yerba
- Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kevin A Porras
- UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Adam M Burgoyne
- Division of Hematology-Oncology, Department of Medicine, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA. .,Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
33
|
Sen I, Young WF, Kasperbauer JL, Polonis K, Harmsen WS, Colglazier JJ, DeMartino RR, Oderich GS, Kalra M, Bower TC. Tumor-specific prognosis of mutation-positive patients with head and neck paragangliomas. J Vasc Surg 2020; 71:1602-1612.e2. [PMID: 32035780 DOI: 10.1016/j.jvs.2019.08.232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Genetic testing to identify succinate dehydrogenase (SDH) mutations in patients with head and neck paraganglioma (HNP) has been in clinical practice for more than a decade. However, the recurrence and metachronous tumor occurrence risks in surgically treated mutation-positive patients are not well studied. METHODS Clinical and procedural details of consecutive patients who underwent excision for HNP from January 1996 to October 2016 were retrospectively reviewed. End points included recurrence, metachronous tumor detection, and mortality. Germline DNA was tested to identify mutations in SDHx genes. Patients were divided into three groups on the basis of genetic testing: group I, positive; group II, negative; and group III, unknown or offered but not tested. RESULTS HNP was diagnosed in 268 patients, 214 (147 female; mean age, 47 years) included in this study. Directed genetic testing was performed in 68; mutations were detected in SDH in 47 (69%), a majority SDHD. In group I, 47 patients had 64 procedures for 81 tumors (52 carotid body tumors [CBTs]); 17 (36%) were bilateral, 7 (15%) multiple, 3 (6%) functional, and 7 (15%) malignant. Residual tumor in 10 was significant in 2, managed by radiation therapy and reoperation. Local recurrence was detected in 12 patients (25%) at a median of 8 years; 11 metachronous mediastinal and retroperitoneal paragangliomas were detected in 8 (17%) at a median of 13 years. Systemic metastases occurred in five (10%). Six patients (13%) had more than one recurrence. In group II, 21 patients had 22 procedures for 23 tumors, 17 CBTs. Two (9%) were bilateral and two (9%) malignant. Excision was complete in all with no recurrence or systemic metastasis at last follow-up. For group III, 146 patients underwent 153 procedures for 156 tumors, 95 CBTs; 7 (5%) were bilateral, 2 (1%) multiple, 8 (5%) functional, and 1 (0.6%) malignant. Local recurrence was detected in nine (6%) at a median of 9 years and metachronous HNP in three (2%) at a median of 5 years. Systemic metastases occurred in two (1%). Mortality was 4% in group I and 3% in group III, none procedure or tumor related. Group I (mutation positive) had 10-year overall, recurrence-free, and metachronous tumor-free survival rates of 93%, 69.4%, and 73%, respectively, lower than the other groups (P < .001). CONCLUSIONS Bilateral, functional, malignant, recurrent, and metachronous tumors are more common in SDH mutation-positive patients with HNP. Overall survival in patients with HNP is high. Metachronous tumors or local recurrences occur late, and long-term follow-up is necessary.
Collapse
Affiliation(s)
- Indrani Sen
- Department of Vascular Surgery, Mayo Clinic, Rochester, Minn.
| | | | | | | | - William S Harmsen
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minn
| | | | | | | | - Manju Kalra
- Department of Vascular Surgery, Mayo Clinic, Rochester, Minn
| | - Thomas C Bower
- Department of Vascular Surgery, Mayo Clinic, Rochester, Minn
| |
Collapse
|
34
|
Marquez J, Flores J, Kim AH, Nyamaa B, Nguyen ATT, Park N, Han J. Rescue of TCA Cycle Dysfunction for Cancer Therapy. J Clin Med 2019; 8:jcm8122161. [PMID: 31817761 PMCID: PMC6947145 DOI: 10.3390/jcm8122161] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 11/30/2019] [Accepted: 12/04/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondrion, a maternally hereditary, subcellular organelle, is the site of the tricarboxylic acid (TCA) cycle, electron transport chain (ETC), and oxidative phosphorylation (OXPHOS)—the basic processes of ATP production. Mitochondrial function plays a pivotal role in the development and pathology of different cancers. Disruption in its activity, like mutations in its TCA cycle enzymes, leads to physiological imbalances and metabolic shifts of the cell, which contributes to the progression of cancer. In this review, we explored the different significant mutations in the mitochondrial enzymes participating in the TCA cycle and the diseases, especially cancer types, that these malfunctions are closely associated with. In addition, this paper also discussed the different therapeutic approaches which are currently being developed to address these diseases caused by mitochondrial enzyme malfunction.
Collapse
Affiliation(s)
- Jubert Marquez
- Department of Health Science and Technology, College of Medicine, Inje University, Busan 47392, Korea; (J.M.); (A.H.K.)
| | - Jessa Flores
- Department of Physiology, College of Medicine, Inje University, Busan 47392, Korea; (J.F.); (B.N.); (A.T.T.N.)
| | - Amy Hyein Kim
- Department of Health Science and Technology, College of Medicine, Inje University, Busan 47392, Korea; (J.M.); (A.H.K.)
| | - Bayalagmaa Nyamaa
- Department of Physiology, College of Medicine, Inje University, Busan 47392, Korea; (J.F.); (B.N.); (A.T.T.N.)
- Department of Hematology, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia
| | - Anh Thi Tuyet Nguyen
- Department of Physiology, College of Medicine, Inje University, Busan 47392, Korea; (J.F.); (B.N.); (A.T.T.N.)
| | - Nammi Park
- Cardiovascular and Metabolic Disease Center, Paik Hospital, Inje University, Busan 47392, Korea;
| | - Jin Han
- Department of Health Science and Technology, College of Medicine, Inje University, Busan 47392, Korea; (J.M.); (A.H.K.)
- Department of Physiology, College of Medicine, Inje University, Busan 47392, Korea; (J.F.); (B.N.); (A.T.T.N.)
- Cardiovascular and Metabolic Disease Center, Paik Hospital, Inje University, Busan 47392, Korea;
- Correspondence: ; Tel.: +8251-890-8748
| |
Collapse
|
35
|
Fankhauser M, Bechmann N, Lauseker M, Goncalves J, Favier J, Klink B, William D, Gieldon L, Maurer J, Spöttl G, Rank P, Knösel T, Orth M, Ziegler CG, Aristizabal Prada ET, Rubinstein G, Fassnacht M, Spitzweg C, Grossman AB, Pacak K, Beuschlein F, Bornstein SR, Eisenhofer G, Auernhammer CJ, Reincke M, Nölting S. Synergistic Highly Potent Targeted Drug Combinations in Different Pheochromocytoma Models Including Human Tumor Cultures. Endocrinology 2019; 160:2600-2617. [PMID: 31322702 PMCID: PMC6795182 DOI: 10.1210/en.2019-00410] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/14/2019] [Indexed: 01/09/2023]
Abstract
There are no officially approved therapies for metastatic pheochromocytomas apart from ultratrace 131I-metaiodbenzylguanidine therapy, which is approved only in the United States. We have, therefore, investigated the antitumor potential of molecular-targeted approaches in murine pheochromocytoma cell lines [monocyte chemoattractant protein (MPC)/monocyte chemoattractant protein/3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], immortalized mouse chromaffin Sdhb-/- cells, three-dimensional pheochromocytoma tumor models (MPC/MTT spheroids), and human pheochromocytoma primary cultures. We identified the specific phosphatidylinositol-3-kinase α inhibitor BYL719 and the mammalian target of rapamycin inhibitor everolimus as the most effective combination in all models. Single treatment with clinically relevant doses of BYL719 and everolimus significantly decreased MPC/MTT and Sdhb-/- cell viability. A targeted combination of both inhibitors synergistically reduced MPC and Sdhb-/- cell viability and showed an additive effect on MTT cells. In MPC/MTT spheroids, treatment with clinically relevant doses of BYL719 alone or in combination with everolimus was highly effective, leading to a significant shrinkage or even a complete collapse of the spheroids. We confirmed the synergism of clinically relevant doses of BYL719 plus everolimus in human pheochromocytoma primary cultures of individual patient tumors with BYL719 attenuating everolimus-induced AKT activation. We have thus established a method to assess molecular-targeted therapies in human pheochromocytoma cultures and identified a highly effective combination therapy. Our data pave the way to customized combination therapy to target individual patient tumors.
Collapse
Affiliation(s)
- Maria Fankhauser
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Michael Lauseker
- Institute for Medical Information Sciences, Biometry, and Epidemiology, Campus Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Judith Goncalves
- Institut National de la Santé et de la Recherche Médicale, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Judith Favier
- Institut National de la Santé et de la Recherche Médicale, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center of Genetics, Laboratoire National de Santé, Dudelange, Luxembourg
- German Cancer Consortium, Dresden, Germany
| | | | - Laura Gieldon
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium, Dresden, Germany
- German Cancer Research Center, Heidelberg, Germany
- Core Unit for Molecular Tumor Diagnostics, National Center for Tumor Diseases, Heidelberg, Germany
| | - Julian Maurer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Gerald Spöttl
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Petra Rank
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Christian G Ziegler
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | | | - German Rubinstein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Martin Fassnacht
- Department of Medicine I, Division of Endocrinology and Diabetology, University Hospital, University of Würzburg, Würzburg, Germany
| | - Christine Spitzweg
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Ashley B Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Royal Free Hospital ENETS Centre of Excellence, London, United Kingdom
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Christoph J Auernhammer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
| | - Svenja Nölting
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU München, Munich, Germany
- Correspondence: Svenja Nölting, MD, Med. Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstraße 1, 80336 Munich, Germany. E-mail:
| |
Collapse
|
36
|
Nölting S, Ullrich M, Pietzsch J, Ziegler CG, Eisenhofer G, Grossman A, Pacak K. Current Management of Pheochromocytoma/Paraganglioma: A Guide for the Practicing Clinician in the Era of Precision Medicine. Cancers (Basel) 2019; 11:cancers11101505. [PMID: 31597347 PMCID: PMC6827093 DOI: 10.3390/cancers11101505] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/18/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PCC/PGLs) are rare, mostly catecholamine-producing neuroendocrine tumors of the adrenal gland (PCCs) or the extra-adrenal paraganglia (PGL). They can be separated into three different molecular clusters depending on their underlying gene mutations in any of the at least 20 known susceptibility genes: The pseudohypoxia-associated cluster 1, the kinase signaling-associated cluster 2, and the Wnt signaling-associated cluster 3. In addition to tumor size, location (adrenal vs. extra-adrenal), multiplicity, age of first diagnosis, and presence of metastatic disease (including tumor burden), other decisive factors for best clinical management of PCC/PGL include the underlying germline mutation. The above factors can impact the choice of different biomarkers and imaging modalities for PCC/PGL diagnosis, as well as screening for other neoplasms, staging, follow-up, and therapy options. This review provides a guide for practicing clinicians summarizing current management of PCC/PGL according to tumor size, location, age of first diagnosis, presence of metastases, and especially underlying mutations in the era of precision medicine.
Collapse
Affiliation(s)
- Svenja Nölting
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstraße 1, 80336 München, Germany.
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
- Department of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstrasse 9, 01062 Dresden, Germany.
| | - Christian G Ziegler
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Graeme Eisenhofer
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, 01307 Dresden, Germany.
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford Ox3 7LJ, UK.
- Department of Gastroenterology, Royal Free Hospital ENETS Centre of Excellence, London NW3 2QG, UK.
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA.
| |
Collapse
|
37
|
Gómez AM, Soares DC, Costa AAB, Pereira DP, Achatz MI, Formiga MN. Pheochromocytoma and paraganglioma: implications of germline mutation investigation for treatment, screening, and surveillance. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:369-375. [PMID: 31365623 PMCID: PMC10528659 DOI: 10.20945/2359-3997000000145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/18/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Paraganglioma (PGL) and pheochromocytoma (PCC) are rare neuroendocrine tumors that were considered to be predominantly sporadic. However, with the identification of novel susceptibility genes over the last decade, it is currently estimated that up to 40% of cases can occur in the context of a hereditary syndrome. We aimed to characterize PGL/PCC families to exemplify the different scenarios in which hereditary syndromes can be suspected and to emphasize the importance for patients and their families of making an opportune genetic diagnosis. MATERIALS AND METHODS Retrospective analysis of patients diagnosed with PGL/PCC. Germline mutations were studied using next-generation sequencing panels including SDHA, SDHB, SDHC and SDHD. Clinical data were collected from clinical records, and all patients received genetic counseling. RESULTS We describe 4 families with PGL/PCC and germline mutations in SDH complex genes. 2 families have SDHB mutations and 2 SDHD mutations. The clinical presentation of the patients and their families was heterogeneous, with some being atypical according to the literature. CONCLUSIONS PGL/PCC are more commonly associated with a germline mutation than any other cancer type, therefore, all individuals with these types of tumors should undergo genetic risk evaluation. NGS multigene panel testing is a cost-effective approach given the overlapping phenotypes. Individuals with germline mutations associated with PGL/PCC should undergo lifelong clinical, biochemical and imaging surveillance and their families should undergo genetic counseling. For all these reasons, it is critical that all medical staff can suspect and diagnose these inherited cancer predisposition syndromes.
Collapse
Affiliation(s)
- Ana Milena Gómez
- Hospital Universitario San IgnacioBogotáColombiaHospital Universitario San Ignacio, Bogotá, Colombia
| | - Diogo Cordeiro Soares
- Departamento de OncogenéticaA.C. Camargo Cancer CenterSão PauloSPBrasilDepartamento de Oncogenética, A.C. Camargo Cancer Center, São Paulo, SP, Brasil
| | - Alexandre André Balieiro Costa
- Departamento de OncogenéticaA.C. Camargo Cancer CenterSão PauloSPBrasilDepartamento de Oncogenética, A.C. Camargo Cancer Center, São Paulo, SP, Brasil
| | - Daniele Paixão Pereira
- Departamento de OncogenéticaA.C. Camargo Cancer CenterSão PauloSPBrasilDepartamento de Oncogenética, A.C. Camargo Cancer Center, São Paulo, SP, Brasil
| | - Maria Isabel Achatz
- Hospital Sírio-LibanêsCentro de OncologiaHospital Sírio-LibanêsSão PauloSPBrasilCentro de Oncologia, Hospital Sírio-Libanês, São Paulo, SP, Brasil
| | - Maria Nirvana Formiga
- Departamento de OncogenéticaA.C. Camargo Cancer CenterSão PauloSPBrasilDepartamento de Oncogenética, A.C. Camargo Cancer Center, São Paulo, SP, Brasil
| |
Collapse
|
38
|
Abstract
Since Felix Fränkel's account of pheochromocytoma in 1886, great discoveries and vast advancements in the diagnosis, genetics, anatomical and functional imaging techniques, and surgical management of pheochromcytoma and paraganglioma (P-PGL) have been made. The improved insight in the pathophysiology of P-PGL and more accurate detection methods enable physicians to tailor the treatment plan to an individual based on the genetic profile and tumor behavior. This review will cover briefly the clinical features, diagnosis, genetic mutations, and imaging modalities that are used to guide current surgical management of these rare and interesting endocrinopathies.
Collapse
Affiliation(s)
- Douglas Wiseman
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mustapha El Lakis
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
39
|
Abstract
Carney-Stratakis Syndrome (CSS) comprises of paragangliomas (PGLs) and gastrointestinal stromal tumors (GISTs). Several of its features overlap with Carney Triad (CT) - PGLs, GISTs, and pulmonary chondromas. CSS has autosomal dominant inheritance, incomplete penetrance, and greater relative frequency of PGL over GISTs. The PGLs in CSS are multicentric and GISTs are multifocal in all the patients, suggesting an inherited susceptibility and associating the two manifestations. In this review, we highlight the clinical, pathological, and molecular characteristics of CSS, along with its diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Arushi Khurana
- VCU Massey Cancer Center - Hematology Oncology, Richmond, Virginia, USA
| | - Lin Mei
- VCU Massey Cancer Center - Hematology Oncology, Richmond, Virginia, USA
| | - Anthony C Faber
- Virginia Commonwealth University - Philips Institute for Oral Health Research, Richmond, Virginia, USA
| | - Steven C Smith
- Virginia Commonwealth University - Pathology, Richmond, Virginia, USA
| | | |
Collapse
|
40
|
Kolasinska-Ćwikła A, Pęczkowska M, Ćwikła JB, Michałowska I, Pałucki JM, Bodei L, Lewczuk-Myślicka A, Januszewicz A. A Clinical Efficacy of PRRT in Patients with Advanced, Nonresectable, Paraganglioma-Pheochromocytoma, Related to SDHx Gene Mutation. J Clin Med 2019; 8:jcm8070952. [PMID: 31262070 PMCID: PMC6678858 DOI: 10.3390/jcm8070952] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
Paragangliomas and pheochromytomas (PPGLs) exhibit variable malignancy, advanced/hormonally active/progressive need therapy. PRRT (Peptide Receptor Radionuclide Therapy) could be an option for these patients. To evaluate the effectiveness of PRRT (90Y DOTATATE), based on overall survival (OS) and progression-free survival (PFS), in patients with PPGLs, related to SDHx gene mutation, we conducted a prospective open-label, single-center, phase II study. Thirteen patients were observed, eight PGL1 and five PGL4, all with advanced, non-resectable tumors, and eight had metastases. All were treated with 90Y DOTATATE. Efficacy was based on OS and PFS, and radiological response was based on RECIST. Hormonal activity was evaluated using serum-fractionated free catecholamines. Eight subjects had a clinical response, three were stable, and two exhibited disease progression. Among four patients with hormonally-active PPGLs, three showed a reduction and one showed normalization. OS for all was 68.0 months; PFS was 35.0 months. OS in PGL4 = 25.0 vs. N.R. (not reached) in PGL1. PFS in PGL4 = 12.0 vs. N.R. in PGL1. A difference was seen in the OS and PFS in patients who did not respond clinically, compared to those who did, OS = 22.0 vs. N.R. PFS = 7.0 vs. N.R. A difference in the OS and PFS was noted in patients with liver and bone involvement compared to those without. PRRT is an effective therapy in selected population of patients with SDHx, in those with locally-advanced, non-resectable tumors. Furthermore, it is effective regardless of the secretory status.
Collapse
Affiliation(s)
- Agnieszka Kolasinska-Ćwikła
- Department of Oncology and Radiotherapy and Department of Radiology, Maria Skłodowska-Curie Memorial Cancer Center, 02-034 Warsaw, Poland
| | - Mariola Pęczkowska
- Department of Hypertension and Department of Radiology Institute of Cardiology, 04-628 Warsaw, Poland
| | - Jarosław B Ćwikła
- Department of Cardiology and Cardiosurgery, School of Medicine; University of Warmia and Mazury, 10-082 Olsztyn, Poland.
| | - Ilona Michałowska
- Department of Hypertension and Department of Radiology Institute of Cardiology, 04-628 Warsaw, Poland
| | - Jakub M Pałucki
- Department of Oncology and Radiotherapy and Department of Radiology, Maria Skłodowska-Curie Memorial Cancer Center, 02-034 Warsaw, Poland
| | - Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Lewczuk-Myślicka
- Department of Internal Medicine and Endocrinology, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Andrzej Januszewicz
- Department of Hypertension and Department of Radiology Institute of Cardiology, 04-628 Warsaw, Poland
| |
Collapse
|
41
|
Performance of plasma free metanephrines in diagnosis of pheochromocytomas and paragangliomas in the population of Asturias. ENDOCRINOLOGÍA, DIABETES Y NUTRICIÓN (ENGLISH ED.) 2019. [DOI: 10.1016/j.endien.2018.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
42
|
Martínez-Morillo E, Valdés Gallego N, Eguia Ángeles E, Fernández Fernández JC, Prieto García B, Álvarez FV. Rendimiento de las metanefrinas libres plasmáticas en el diagnóstico de los feocromocitomas y paragangliomas en la población asturiana. ENDOCRINOL DIAB NUTR 2019; 66:312-319. [DOI: 10.1016/j.endinu.2018.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 11/30/2022]
|
43
|
Molecular Alterations in Dog Pheochromocytomas and Paragangliomas. Cancers (Basel) 2019; 11:cancers11050607. [PMID: 31052272 PMCID: PMC6563419 DOI: 10.3390/cancers11050607] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022] Open
Abstract
8658860258318000Recently, genetic alterations in the genes encoding succinate dehydrogenase subunit B and D (SDHB and SDHD) were identified in pet dogs that presented with spontaneously arising pheochromocytomas (PCC) and paragangliomas (PGL; together PPGL), suggesting dogs might be an interesting comparative model for the study of human PPGL. To study whether canine PPGL resembled human PPGL, we investigated a series of 50 canine PPGLs by immunohistochemistry to determine the expression of synaptophysin (SYP), tyrosine hydroxylase (TH) and succinate dehydrogenase subunit A (SDHA) and B (SDHB). In parallel, 25 canine PPGLs were screened for mutations in SDHB and SDHD by Sanger sequencing. To detect large chromosomal alterations, single nucleotide polymorphism (SNP) arrays were performed for 11 PPGLs, including cases for which fresh frozen tissue was available. The immunohistochemical markers stained positive in the majority of canine PPGLs. Genetic screening of the canine tumors revealed the previously described variants in four cases; SDHB p.Arg38Gln (n = 1) and SDHD p.Lys122Arg (n = 3). Furthermore, the SNP arrays revealed large chromosomal alterations of which the loss of chromosome 5, partly homologous to human chromosome 1p and chromosome 11, was the most frequent finding (100% of the six cases with chromosomal alterations). In conclusion, canine and human PPGLs show similar genomic alterations, suggestive of common interspecies PPGL-related pathways.
Collapse
|
44
|
Wong MY, Andrews KA, Challis BG, Park S, Acerini CL, Maher ER, Casey RT. Clinical Practice Guidance: Surveillance for phaeochromocytoma and paraganglioma in paediatric succinate dehydrogenase gene mutation carriers. Clin Endocrinol (Oxf) 2019; 90:499-505. [PMID: 30589099 PMCID: PMC6850004 DOI: 10.1111/cen.13926] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/19/2018] [Accepted: 12/23/2018] [Indexed: 01/08/2023]
Abstract
The succinate dehydrogenase (SDH) enzyme complex functions as a key enzyme coupling the oxidation of succinate to fumarate in the citric acid cycle. Inactivation of this enzyme complex results in the cellular accumulation of the oncometabolite succinate, which is postulated to be a key driver in tumorigenesis. Succinate accumulation inhibits 2-oxoglutarate-dependent dioxygenases, including DNA and histone demethylase enzymes and hypoxic gene response regulators. Biallelic inactivation (typically resulting from one inherited and one somatic event) at one of the four genes encoding the SDH complex (SDHA/B/C/D) is the most common cause for SDH deficient (dSDH) tumours. Germline mutations in the SDHx genes predispose to a spectrum of tumours including phaeochromocytoma and paraganglioma (PPGL), wild type gastrointestinal stromal tumours (wtGIST) and, less commonly, renal cell carcinoma and pituitary tumours. Furthermore, mutations in the SDHx genes, particularly SDHB, predispose to a higher risk of malignant PPGL, which is associated with a 5-year mortality of 50%. There is general agreement that biochemical and imaging surveillance should be offered to asymptomatic carriers of SDHx gene mutations in the expectation that this will reduce the morbidity and mortality associated with dSDH tumours. However, there is no consensus on when and how surveillance should be performed in children and young adults. Here, we address the question: "What age should clinical, biochemical and radiological surveillance for PPGL be initiated in paediatric SDHx mutation carriers?".
Collapse
Affiliation(s)
- Mei Yin Wong
- Department of Diabetes and EndocrinologyCambridge University Hospital NHS Foundation TrustCambridgeUK
| | - Katrina A. Andrews
- East Anglian Medical Genetics ServiceCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Benjamin G. Challis
- Department of Diabetes and EndocrinologyCambridge University Hospital NHS Foundation TrustCambridgeUK
| | - Soo‐Mi Park
- East Anglian Medical Genetics ServiceCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | | | - Eamonn R. Maher
- Department of Medical GeneticsUniversity of CambridgeCambridgeUK
- NIHR Cambridge Biomedical Research Centre and Cancer Research UK Cambridge CentreCambridgeUK
| | - Ruth T. Casey
- Department of Diabetes and EndocrinologyCambridge University Hospital NHS Foundation TrustCambridgeUK
- Department of Medical GeneticsUniversity of CambridgeCambridgeUK
| |
Collapse
|
45
|
Pheochromocytomas and Paragangliomas: From Genetic Diversity to Targeted Therapies. Cancers (Basel) 2019; 11:cancers11040436. [PMID: 30925729 PMCID: PMC6521122 DOI: 10.3390/cancers11040436] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Pheochromocytoma and paraganglioma (PCPGs) are rare neuroendocrine tumors that arise from the chromaffin tissue of adrenal medulla and sympathetic ganglia. Although metastatic PCPGs account for only 10% of clinical cases, morbidity and mortality are high because of the uncontrollable mass effect and catecholamine level generated by these tumors. Despite our expanding knowledge of PCPG genetics, the clinical options to effectively suppress PCPG progression remain limited. Several recent translational studies revealed that PCPGs with different molecular subtypes exhibit distinctive oncogenic pathways and spectrum of therapy resistance. This suggests that therapeutics can be adjusted based on the signature molecular and metabolic pathways of PCPGs. In this review, we summarized the latest findings on PCPG genetics, novel therapeutic targets, and perspectives for future personalized medicine.
Collapse
|
46
|
Tufton N, Shapiro L, Sahdev A, Kumar AV, Martin L, Drake WM, Akker SA, Storr HL. An analysis of surveillance screening for SDHB-related disease in childhood and adolescence. Endocr Connect 2019; 8:162-172. [PMID: 30694796 PMCID: PMC6391899 DOI: 10.1530/ec-18-0522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 11/26/2022]
Abstract
Objective Phaeochromocytomas (PCC) and paragangliomas (PGL) are rare in children. A large proportion of these are now understood to be due to underlying germline mutations. Here we focus on succinate dehydrogenase subunit B (SDHB) gene mutation carriers as these tumours carry a high risk of malignant transformation. There remains no current consensus with respect to optimal surveillance for asymptomatic carriers and those in whom the presenting tumour has been resected. Method We undertook a retrospective analysis of longitudinal clinical data of all children and adolescents with SDHB mutations followed up in a single UK tertiary referral centre. This included index cases that pre-dated the introduction of surveillance screening and asymptomatic carriers identified through cascade genetic testing. We also conducted a literature review to inform a suggested surveillance protocol for children and adolescents harbouring SDHB mutations. Results Clinical outcomes of a total of 38 children are presented: 8 index cases and 30 mutation-positive asymptomatic carriers with 175 patient years of follow-up data. Three of the eight index cases developed metachronous disease and two developed metastatic disease. Of the 30 asymptomatic carriers, 3 were found to have PGLs on surveillance screening. Conclusions Surveillance screening was well tolerated in our paediatric cohort and asymptomatic paediatric subjects. Screening can identify tumours before they become secretory and/or symptomatic, thereby facilitating surgical resection and reducing the chance of distant spread. We propose a regular screening protocol commencing at age 5 years in this at-risk cohort of patients.
Collapse
Affiliation(s)
- Nicola Tufton
- Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Lucy Shapiro
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Paediatric Endocrinology, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Anju Sahdev
- Department of Radiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
| | - Ajith V Kumar
- North East Thames Regional Genetics Service, Great Ormond Street Hospital, London, UK
| | - Lee Martin
- Department of Paediatric Endocrinology, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - William M Drake
- Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Scott A Akker
- Department of Endocrinology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helen L Storr
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Paediatric Endocrinology, Royal London Hospital, Barts Health NHS Trust, London, UK
- Correspondence should be addressed to H L Storr:
| |
Collapse
|
47
|
Increased Mortality in SDHB but Not in SDHD Pathogenic Variant Carriers. Cancers (Basel) 2019; 11:cancers11010103. [PMID: 30658386 PMCID: PMC6356820 DOI: 10.3390/cancers11010103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Germline mutations in succinate dehydrogenase subunit B and D (SDHB and SDHD) are predisposed to hereditary paraganglioma (PGL) and pheochromocytoma (PHEO). The phenotype of pathogenic variants varies according to the causative gene. In this retrospective study, we estimate the mortality of a nationwide cohort of SDHB variant carriers and that of a large cohort of SDHD variant carriers and compare it to the mortality of a matched cohort of the general Dutch population. A total of 192 SDHB variant carriers and 232 SDHD variant carriers were included in this study. The Standard Mortality Ratio (SMR) for SDHB mutation carriers was 1.89, increasing to 2.88 in carriers affected by PGL. For SDHD variant carriers the SMR was 0.93 and 1.06 in affected carriers. Compared to the general population, mortality seems to be increased in SDHB variant carriers, especially in those affected by PGL. In SDHD variant carriers, the mortality is comparable to that of the general Dutch population, even if they are affected by PGL. This insight emphasizes the significance of DNA-testing in all PGL and PHEO patients, since different clinical risks may warrant gene-specific management strategies.
Collapse
|
48
|
Blefari ND, Eliezer DD, Vilain RE, Hesketh EE, Deshpande A, Deshpande AV. Sclerosing paraganglioma of the carotid body in a child: A diagnostic and therapeutic challenge. INT J PEDIAT OTO CAS 2019. [DOI: 10.1016/j.pedeo.2019.100663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Tufton N, Sahdev A, Drake WM, Akker SA. Can subunit-specific phenotypes guide surveillance imaging decisions in asymptomatic SDH mutation carriers? Clin Endocrinol (Oxf) 2019; 90:31-46. [PMID: 30303539 DOI: 10.1111/cen.13877] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/07/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE With the discovery that familial phaeochromocytoma and paraganglioma syndrome can be caused by mutations in each subunit of the succinate dehydrogenase enzyme (SDH), has come the recognition that mutations in the individual subunits have their own distinct natural histories. Increased genetic screening is leading to the identification of increasing numbers of, mostly asymptomatic, gene mutation carriers and the implementation of screening strategies for these individuals. Yet there is, to date, no international consensus regarding screening strategies for asymptomatic carriers. DESIGN A comprehensive PubMed search from 1/1/2000 to 28/2/2018 was undertaken using multiple search terms and subsequently a manual review of references in identified papers to identify all clinically relevant cases and cohorts. In this review, the accumulated, published experience of phenotype and malignancy risks of individual SDH subunits is analysed. Where possible screening results for asymptomatic SDH mutation carriers have been analysed separately to define the penetrance in asymptomatic carriers (asymptomatic penetrance). RESULTS The combined data confirms that "asymptomatic penetrance" is highest for SDHD and when there is penetrance, the most likely site to develop a PGL is head and neck (SDHD) and extra-adrenal abdominal (SDHB). However, the risk in SDHB carriers of developing HNPGL is also high (35.5%) and a PCC is low (15.1%), and in SDHD carriers there is a high risk of developing a PCC (35.8%) or abdominal PGL (9.4%) and a small, but significant risk at other sympathetic sites. The data suggest that the risk of malignant transformation is the same for both PCC and extra-adrenal abdominal PGLs (30%-35%) in SDHB carriers. In SDHD carriers, the risk of malignant transformation was highest in HNPGLs (7.5%) and similar for sympathetic sites (3.8%-5.2%). CONCLUSIONS Using this data, we suggest surveillance screening of asymptomatic carriers can be tailored to the underlying SDH subunit and review possible surveillance programmes.
Collapse
Affiliation(s)
- Nicola Tufton
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anju Sahdev
- Department of Radiology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - William M Drake
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Scott A Akker
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
50
|
Huang Y, Wang LA, Xie Q, Pang J, Wang L, Yi Y, Zhang J, Zhang Y, Chen R, Lan W, Zhang D, Jiang J. Germline SDHB and SDHD mutations in pheochromocytoma and paraganglioma patients. Endocr Connect 2018; 7:1217-1225. [PMID: 30352407 PMCID: PMC6240141 DOI: 10.1530/ec-18-0325] [Citation(s) in RCA: 15] [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: 09/20/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022]
Abstract
Pheochromocytoma and paragangliomas (PCC/PGL) are neuroendocrine tumors that arise from chromaffin cells of the adrenal medulla and sympathetic/parasympathetic ganglia, respectively. Of clinical relevance regarding diagnosis is the highly variable presentation of symptoms in PCC/PGL patients. To date, the clear-cut correlations between the genotypes and phenotypes of PCC/PGL have not been entirely established. In this study, we reviewed the medical records of PCC/PGL patients with pertinent clinical, laboratory and genetic information. Next-generation sequencing (NGS) performed on patient samples revealed specific germline mutations in the SDHB (succinate dehydrogenase complex iron-sulfur subunit B) and SDHD (succinate dehydrogenase complex subunit D) genes and these mutations were validated by Sanger sequencing. Of the 119 patients, two were identified with SDHB mutation and one with SDHD mutation. Immunohistochemical (IHC) staining was used to analyze the expression of these mutated genes. The germline mutations identified in the SDH genes were c343C>T and c.541-542A>G in the SDHB gene and c.334-337delACTG in the SDHD gene. IHC staining of tumors from the c.343C>T and c.541-2A>G carriers showed positive expression of SDHB. Tumors from the c.334-337delACTG carrier showed no expression of SDHD and a weak diffused staining pattern for SDHB. We strongly recommend genetic testing for suspected PCC/PGL patients with a positive family history, early onset of age, erratic hypertension, recurrence or multiple tumor sites and loss of SDHB and/or SDHD expression. Tailored personal management should be conducted once a patient is confirmed as an SDHB and/or SDHD mutation carrier or diagnosed with PCC/PGL.
Collapse
Affiliation(s)
- Yiqiang Huang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Lin-ang Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Qiubo Xie
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jian Pang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Luofu Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yuting Yi
- Geneplus-Beijing Institute, Beijing, People’s Republic of China
| | - Jun Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yao Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Rongrong Chen
- Geneplus-Beijing Institute, Beijing, People’s Republic of China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|