1
|
Zhou Y, Tai Y, Shang J. Progress in treatment and follow-up of pheochromocytoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110144. [PMID: 40373734 DOI: 10.1016/j.ejso.2025.110144] [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: 01/14/2025] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
Pheochromocytoma (PHEO) is an uncommon neuroendocrine tumor originating from the chromaffin cells of the adrenal medulla. These tumors are capable of producing and releasing substantial amounts of catecholamine (CA) hormones. Individuals with PHEO often experience transient blood pressure fluctuations, headaches, and palpitations, among other symptoms. In extreme cases, they may develop severe complications, including cardiovascular and cerebrovascular incidents, myocardial disorders, and gastrointestinal issues. At the advanced stage, PHEO can affect multiple organs, potentially leading to pheochromocytoma crisis. However, the clinical manifestations of pheochromocytoma may be diverse. Some patients have no typical triad of headache, palpitations, and sweating, and are not accompanied by obvious clinical symptoms or signs. There are only abnormalities in imaging and biochemical indicators, which will pose a challenge for early diagnosis. The primary treatment options for PHEO encompass surgical and non-surgical approaches. In contrast to many other adrenal pathologies, there is currently no consensus on the optimal surgical versus non-surgical management of PHEO. Laparoscopic surgery, as opposed to traditional open surgery, offers numerous benefits. However, whether retroperitoneal or transperitoneal laparoscopic adrenalectomy remains controversial. Da Vinci robot-assisted adrenalectomy has the advantages of highly precise operation and excellent hemostasis capabilities. The day surgery management model for adrenalectomy procedures has proven to be both safe and feasible. However, current research on its long - term effectiveness and wide - scale application still has limitations. Additionally, the application of the Senhance robot in urology, particularly in adrenalectomy, has seen relatively little investigation thus far. In terms of non-surgical treatment for pheochromocytoma, chemotherapy, radionuclide therapy, targeted therapy and immunotherapy, as well as radiofrequency ablation and microwave ablation chemotherapy have all advanced significantly. This article aims to review the latest advancements in the treatment of pheochromocytoma.
Collapse
Affiliation(s)
- Yifan Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yanghao Tai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jiwen Shang
- Department of Ambulatory Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
2
|
Arenillas C, Celada L, Ruiz-Cantador J, Calsina B, Datta D, García-Galea E, Fasani R, Moreno-Cárdenas AB, Alba-Linares JJ, Miranda-Barrio B, Martínez-Montes ÁM, Alvarez-Escola C, Lecumberri B, González García A, K. Flores S, Esquivel E, Ding Y, Peitzsch M, Robles-Guirado JÁ, Regojo Zapata RM, Pozo-Kreilinger JJ, Iglesias C, Dwight T, Muir CA, Oleaga A, Garrido-Lestache Rodríguez-Monte ME, Del Cerro MJ, Martínez-Bendayán I, Álvarez-González E, Cubiella T, Lourenço DM, A. Pereira MA, Burnichon N, Buffet A, Broberg C, Dickson PV, Fraga MF, Llorente Pendás JL, Rueda Soriano J, Buendía Fuentes F, Toledo SP, Clifton-Bligh R, Dienstmann R, Villanueva J, Capdevila J, Gimenez-Roqueplo AP, Favier J, Nuciforo P, Young WF, Bechmann N, Opotowsky AR, Vaidya A, Bancos I, Weghorn D, Robledo M, Casteràs A, Dos-Subirà L, Adameyko I, Chiara MD, Dahia PL, Toledo RA. Convergent Genetic Adaptation in Human Tumors Developed Under Systemic Hypoxia and in Populations Living at High Altitudes. Cancer Discov 2025; 15:1037-1062. [PMID: 40199338 PMCID: PMC12046333 DOI: 10.1158/2159-8290.cd-24-0943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
SIGNIFICANCE This study reveals a broad convergence in genetic adaptation to hypoxia between natural populations and tumors, suggesting that insights from natural populations could enhance our understanding of cancer biology and identify novel therapeutic targets. See related commentary by Lee, p. 875.
Collapse
Affiliation(s)
- Carlota Arenillas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lucía Celada
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - José Ruiz-Cantador
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Debayan Datta
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Eduardo García-Galea
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Roberta Fasani
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ana Belén Moreno-Cárdenas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan José Alba-Linares
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Berta Miranda-Barrio
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Ángel M. Martínez-Montes
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, Hospital Universitario La Paz, Madrid, Spain
| | - Ana González García
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Shahida K. Flores
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Emmanuel Esquivel
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Yanli Ding
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - José-Ángel Robles-Guirado
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | | - Carmela Iglesias
- Department of Pathology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Trisha Dwight
- Cancer Genetics, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
- The University of Sydney, Sydney, Australia
| | - Christopher A. Muir
- Department of Endocrinology, St. Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Amelia Oleaga
- Department of Endocrinology and Nutrition, Hospital Universitario de Basurto, Bilbao, Spain
| | | | - Maria Jesús Del Cerro
- Department of Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Isaac Martínez-Bendayán
- Department of Pediatric Cardiology, Instituto de Investigación Biomédica (Cardiopatía Estructural y Congénita) and Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Enol Álvarez-González
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Department of Functional Biology, Genetic Area, University of Oviedo, Oviedo, Spain
| | - Tamara Cubiella
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Delmar Muniz Lourenço
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Maria Adelaide A. Pereira
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Nelly Burnichon
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Alexandre Buffet
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Craig Broberg
- Adult Congenital Heart Program, Division of Cardiology, Oregon Health and Science University, Portland, Oregon
| | - Paxton V. Dickson
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Mario F. Fraga
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Luis Llorente Pendás
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Joaquín Rueda Soriano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Francisco Buendía Fuentes
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Roderick Clifton-Bligh
- Department of Endocrinology and Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- University of Vic – Central University of Catalonia, Vic, Spain
| | - Josep Villanueva
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jaume Capdevila
- Neuroendocrine and Endocrine Tumor Translational Research Program (NET-VHIO), Vall Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medical Oncology, Gastrointestinal and Endocrine Tumor Unit, Vall d’Hebron Hospital Universitari, Vall d’Hebron Hospital Campus, Barcelona, Spain
| | - Anne-Paule Gimenez-Roqueplo
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Judith Favier
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander R. Opotowsky
- Cincinnati Adult Congenital Heart Disease Program, Heart Institute, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, Ohio
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anand Vaidya
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irina Bancos
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anna Casteràs
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Department of Endocrinology and Nutrition, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Dos-Subirà
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - María-Dolores Chiara
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Patricia L.M. Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Rodrigo A. Toledo
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
3
|
Li Z, Lai D, Jia Y, Luo J, Ma X, Zhang X, Tang L, Li X, Wang B. Predictors of postoperative recurrence of pheochromocytoma: a monocentric study. BMC Surg 2025; 25:179. [PMID: 40281549 PMCID: PMC12023388 DOI: 10.1186/s12893-025-02824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVE To discuss the risk factors affecting the recurrence of pheochromocytoma (PCC) following surgery. METHODS We retrospectively reviewed patients who were hospitalized and underwent surgery for PCC between January 2012 and December 2020 at Chinese People's Liberation Army (PLA) General Hospital. Inclusion criteria were pathological diagnosis of PCC and availability of follow up. RESULTS In total, 451 patients met the inclusion criteria. The average age was 45.89 years, and the median tumor diameter was 5.75 cm. The mean recurrence time was 34.24 months. Of the 451 patients receiving surgery, there were 35 recurrent cases (7.85%). The univariate test showed that age, hypertension, history of PCC recurrence, Ki-67 index ≥ 5, bilateral tumor, duration of phenazopyridine administration, DBP at admission, open operation, intraoperative HR minimum, the number of episodes of intraoperative HR over 120 bpm, the number of episodes of intraoperative hemodynamic instability, and intraoperative bleeding were associated with recurrence after surgery. Multivariate COX regression analysis of age (hazard ratio 0.95), hypertension (hazard ratio 7.14), history of PCC recurrence (hazard ratio 69.35), family history of hypertension (hazard ratio 16.30), bilateral tumor (hazard ratio 7.38), tumor size (hazard ratio 1.05), the number of episodes of hemodynamic instability (hazard ratio 114.91) and duration of intraoperative instability (hazard ratio 1.12) were the independent risk factors on recurrence following surgery. CONCLUSIONS Age, hypertension, history of PCC recurrence, family history of hypertension, bilateral tumor, tumor size, the number of episodes and the duration of intraoperative hemodynamic instability were independent risk factors on recurrence following surgery. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Zhuoran Li
- Medical School of Nankai University, Tianjin, China
| | - Dong Lai
- Department of Urology, Chinese PLA General Hospital/PLA Medical School, Beijing, China
| | - Yuqi Jia
- Medical School of Nankai University, Tianjin, China
| | - Jin Luo
- Medical School of Nankai University, Tianjin, China
| | - Xin Ma
- Department of Urology, Chinese PLA General Hospital/PLA Medical School, Beijing, China
| | - Xu Zhang
- Department of Urology, Chinese PLA General Hospital/PLA Medical School, Beijing, China
| | - Lu Tang
- Department of Urology, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Xintao Li
- Department of Urology, Chinese Air Force Medical Center, Beijing, China.
| | - Baojun Wang
- Department of Urology, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| |
Collapse
|
4
|
Farhat J, Alzyoud L, AlWahsh M, Acharjee A, Al‐Omari B. Advancing Precision Medicine: The Role of Genetic Testing and Sequencing Technologies in Identifying Biological Markers for Rare Cancers. Cancer Med 2025; 14:e70853. [PMID: 40249565 PMCID: PMC12007469 DOI: 10.1002/cam4.70853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/26/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Genetic testing and sequencing technologies offer a comprehensive understanding of cancer genetics, providing rapid and cost-effective solutions. In particular, these advanced technologies play an important role in assessing the complexities of the rare cancer types affecting several systems including the bone, endocrine, digestive, vascular, and soft tissue. This review will explore how genetic testing and sequencing technologies have contributed to the identification of biomarkers across several rare cancer types in diagnostic, therapeutic, and prognostic stages, thereby advancing PM. METHODS A comprehensive literature search was conducted across PubMed (MEDLINE), EMBASE, and Web of Science using keywords related to sequencing technologies, genetic testing, and cancer. There were no restrictions on language, methodology, age, or publication date. Both primary and secondary research involving humans or animals were considered. RESULTS In practice, fluorescence in situ hybridization, karyotype, microarrays and other genetic tests are mainly applied to identify specific genetic alterations and mutations associated with cancer progression. Sequencing technologies, such as next generation sequencing, polymerase chain reaction, whole genome or exome sequencing, enable the rapid analysis of millions of DNA fragments. These techniques assess genome structure, genetic changes, gene expression profiles, and epigenetic variations. Consequently, they help detect main intrinsic markers that are crucial for personalizing diagnosis, treatment options, and prognostic assessments, leading to better patient prognosis. This highlights why these methods are now considered as primary tools in rare cancer research. However, these methods still face multiple limitations, including false positive results, limited precision, and high costs. CONCLUSION Genetic testing and sequencing technologies have significantly advanced the field of rare cancer research by enabling the identification of key biomarkers for precision diagnosis, treatment, and prognosis. Despite existing limitations, their integration into clinical and research fields continues to improve the development of personalized medicine strategies for rare and complex cancer types.
Collapse
Affiliation(s)
- Joviana Farhat
- Department of Epidemiology and Population Health, College of Medicine and Health SciencesKhalifa UniversityAbu DhabiUAE
| | - Lara Alzyoud
- College of PharmacyAl Ain UniversityAbu DhabiUAE
- Health and Biomedical Research CenterAl Ain UniversityAbu DhabiUAE
| | - Mohammad AlWahsh
- Leibniz‐Institut Für Analytische Wissenschaften‐ISAS e.V.DortmundGermany
- Institute of Pathology and Medical Research Center (ZMF) University Medical Center MannheimHeid Elberg UniversityMannheimGermany
- Department of Pharmacy, Faculty of PharmacyAlZaytoonah University of JordanAmmanJordan
| | - Animesh Acharjee
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Basem Al‐Omari
- Department of Epidemiology and Population Health, College of Medicine and Health SciencesKhalifa UniversityAbu DhabiUAE
| |
Collapse
|
5
|
Berti V, Fasciglione E, Charpiot A, Montanini F, Pepponi M, Leo A, Hubele F, Taieb D, Pacak K, Goichot B, Imperiale A. Deciphering 18F-DOPA uptake in SDH-related head and neck paragangliomas: a radiomics approach. J Endocrinol Invest 2025; 48:941-950. [PMID: 39666255 DOI: 10.1007/s40618-024-02515-y] [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: 06/18/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
PURPOSE To investigate the influence of germline succinate dehydrogenase (SDHx) pathogenic variants on 6-[18F]-fluoro-3,4-dihydroxyphenylalanine (18F-DOPA) Positron Emission Tomography (PET) radiomic signature of head and neck paragangliomas (HNPGLs). METHODS Forty-seven patients (20 SDH pathogenic variants carriers) harboring 55 HNPGLs were retrospectively included. HNPGLs were delineated using Nestle adaptive threshold. 128 radiomic features were extracted and harmonized to correct for batch effects. Principal Component Analysis (PCA) was performed to remove redundancy and avoid collinearity. The most representative feature of each component was tested with multivariate stepwise logistic binary regression analysis (LBRA) to identify variables predictive of genetic status. RESULTS 18F-DOPA Positron Emission Tomography/Computed Tomography (PET/CT) detected 28/29 carotid body HNPGLs, 23/23 jugulotympanic HNPGLs, and 4/4 vagal HNPGLs. SUVmax was significantly higher in SDH-related HNPGLs (p = 0.003). PCA allowed identification of 4 Components. The most representative variables of Component 1 and 2 (including intensity and intensity-related textural features, and not intensity-related textural features, respectively) were Intensity-based (IB)-SUVmedian and Grey Level Run Length Matrix-Long Run Low Gray Level Emphasis (GLRLM-LRLGLE). SDHx HNPGLs exhibited higher activity scores and more homogeneous texture. At patient level, SDHx cases showed significantly higher IB-SUVmedian values (p < 0.001), and lower GLRLM-LRLGLE than sporadic patients (p = 0.005). IB-SUVmedian was found to be an independent predictor of genetic status at lesion (71.0%) and patient level (77.8%). CONCLUSION The present study pioneers the application of 18F-DOPA PET radiomics for HNPGLs, suggesting the influence of germline SDH pathogenic variants on 18F-FDOPA uptake intensity and textural heterogeneity. Integrating radiomics with genetic data provides new insights into the correlation between PET features and underlying molecular dysregulation.
Collapse
Affiliation(s)
- Valentina Berti
- Nuclear Medicine Unit, Careggi University Hospital, Florence, Italy
- Experimental and Clinical Biomedical Sciences 'Mario Serio', Florence University, Florence, Italy
| | - Elsa Fasciglione
- Endocrinology, Diabetology, Nutrition, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France
| | - Anne Charpiot
- Otolaryngology and Maxillofacial Surgery, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France
| | - Flavio Montanini
- Nuclear Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Miriam Pepponi
- Nuclear Medicine Unit, Careggi University Hospital, Florence, Italy
- Nuclear Medicine and Molecular Imaging, ICANS, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France
| | - Andrea Leo
- Experimental and Clinical Biomedical Sciences 'Mario Serio', Florence University, Florence, Italy
| | - Fabrice Hubele
- Nuclear Medicine and Molecular Imaging, ICANS, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France
| | - David Taieb
- Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD, USA
| | - Bernard Goichot
- Endocrinology, Diabetology, Nutrition, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, ICANS, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France.
- IPHC, UMR7178, CNRS/Unistra, Strasbourg, France.
- Médecine Nucléaire et Imagerie Moléculaire, ICANS, 17 Rue Albert Calmette, Strasbourg, 67093, France.
| |
Collapse
|
6
|
Jiang J, Pang Y, Luo R, Wei Y, Zhang J, Li M, Xu Y, Teng X, Wu H, Guan H, Wu X, Yan C, Zhong D, Deng W, Xu N, Wen Y, Feng Y, Yan B, Wang L, Jiang Y, Ning J, Xu X, Soria MJ, Robledo M, Pacak K, Liu Y, Liu L. Genetics of urinary bladder paragangliomas: a multi-center study of a Chinese cohort. J Endocrinol Invest 2025; 48:931-939. [PMID: 39636472 DOI: 10.1007/s40618-024-02509-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Pheochromocytomas and paragangliomas (PPGLs) exhibit the highest degree of heritability among all human tumors, yet the genetics of urinary bladder paragangliomas (UBPGLs) remains poorly understood. The present study aims to examine the characteristics of a cohort of Chinese patients with UBPGLs, focusing particularly on genetics. METHODS The study included 70 Chinese patients with UBPGLs from 15 centers in China, 240 patients with non-head and neck PGLs (non-HNPGLs) outside the urine bladder, and 16 Caucasian patients with UBPGLs. Tumor DNA samples were sequenced by next generation sequencing. All identified pathogenic variants (PVs) were confirmed by Sanger sequencing. RESULTS Among the 70 Chinese patients, PVs were identified in 38 cases: 23 in cluster 1 A (13 SDHB, 1 SDHD, 1 SDHA, 4 IDH1, 2 SLC25A11, and 2 FH), 4 in cluster 1B (3 EPAS1 and 1 EGLN1), and 11 in cluster 2 genes (7 HRAS, 1 FGFR1, 2 NF1, and 1 H3F3A). Compared with other non-HNPGLs, UBPGLs had more PVs in cluster 1 A genes (32.9% vs. 14.2%, p < 0.001), but fewer in cluster 1B (5.7% vs. 19.2%, p = 0.002) and cluster 2 genes (15.7% vs. 42.5%, p < 0.001). PVs in SDHB (18.6%) was the most common in Chinese patients with UBPGLs, followed by HRAS (10.0%). No PVs was found in 45.7% of all UBPGLs. PVs in HRAS, SLC25A11, EPAS1, and FH were also identified in Caucasians with UBPGLs. CONCLUSION Chinese patients with UBPGLs have a diverse genetic profile. PVs in cluster 1 A genes underlie nearly 1/3 of patients, highlighting the importance of genetic testing. Diverse germline and somatic PVs are also present in Caucasian patients with UBPGLs.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yongbao Wei
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Jing Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Minghao Li
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yitong Xu
- Department of Pathology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaohong Wu
- Geriatric Medicine Center, Cancer Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Chenyan Yan
- Geriatric Medicine Center, Cancer Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Dewen Zhong
- Department of Urology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Wanglong Deng
- Department of Urology, the First People's Hospital of Chenzhou, Chenzhou, China
| | - Ning Xu
- Department of Urology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yanlin Wen
- Department of Urology, Nanchong Central Hospital, the Second Clinical Hospital of North Sichuan Medical College, Szechwan, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Yan
- Department of Urology, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yazhuo Jiang
- Department of Urology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaowen Xu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Miguel J Soria
- Department of Medical Oncology, European University of Madrid, University Getafe Hospital, Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, Spanish National Cancer Research Center, Madrid, Spain
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road Xuhui District, Shanghai, 200032, China.
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
de Bresser CJM, Rijken JA, van Treijen MJC, van Nesselrooij BPM, de Ridder M, de Bree R, de Borst GJ, Petri BJ, van Leeuwaarde RS. Low psychosocial burden in patients with paraganglioma syndrome: results from the Head and Neck Paraganglioma Registry in a single center. Eur J Endocrinol 2025; 192:257-265. [PMID: 39993160 DOI: 10.1093/ejendo/lvaf033] [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: 11/06/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE Autosomal dominant variants in the succinate dehydrogenase gene (SDHx) are responsible for ∼50% of the development of hereditary paragangliomas and pheochromocytomas (PPGLs). Limited research has been conducted on the psychosocial impact of possessing a hereditary tumor syndrome. In this study, the psychological impact of harboring a genetic variant associated with familial paraganglioma syndrome was assessed. Secondary objectives included the analysis of potential variations in quality of life in (pre)symptomatic stage and comparison with the general Dutch population and other hereditary tumor syndromes. METHODS The first 100 patients from the Head and Neck PGL Registry in the University Medical Center Utrecht were selected. Psychosocial outcomes were assessed cross-sectionally using 5 validated health-related questionnaires: EuroQol 5D-5L, Cancer Worry Scale, Hospital Anxiety and Depression Scale, Modified Fatigue Impact Scale, and EORTC QLQ-C30. RESULTS No significant differences were observed when stratified for (pre)symptomatic status or genetic variant status. Hereditary PPGLs tended to express greater concern about the development of PPGLs in family members. Complaints in the physical domains were more frequently observed in the sporadic group. The PPGL cohort demonstrated better outcomes when compared to other hereditary tumor syndromes and aligned with the Dutch tariff. CONCLUSION The psychosocial impact of harboring a PPGL seems to align with the general healthy Dutch population. Clinical care management involving a multidisciplinary approach and comprehensive counseling on PPGLs and their genetic origins, effectively supports patients. Routine psychological support in the care for these patients does not seem imperative and should be offered indicated on a case-by-case basis.
Collapse
Affiliation(s)
- Carolijn J M de Bresser
- Department of Vascular Surgery, University Utrecht Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Johannes A Rijken
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Mark J C van Treijen
- Department of Endocrine Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht The Netherlands
| | - Bernadette P M van Nesselrooij
- Department of Clinical Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Mischa de Ridder
- Department of Radiotherapy, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery, University Utrecht Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Bart-Jeroen Petri
- Department of Vascular Surgery, University Utrecht Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Rachel S van Leeuwaarde
- Department of Endocrine Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht The Netherlands
| |
Collapse
|
8
|
Gild ML, Do K, Tsang VHM, Tacon LJ, Clifton-Bligh RJ, Robinson BG. Pheochromocytoma in MEN2. Recent Results Cancer Res 2025; 223:211-235. [PMID: 40102259 DOI: 10.1007/978-3-031-80396-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Pheochromocytomas (PCs) are rare neuroendocrine tumors found in 20-50% of MEN2 patients. MEN2-related PCs are more often bilateral, identified at a younger age and have a low metastatic potential. They secrete epinephrine as the predominant catecholamine, along with its metabolite metanephrine, and lesser amounts of norepinephrine and normetanephrine. The advent of molecular diagnostic tools has enhanced the identification and stratification of these tumors, revealing a strong genotype-phenotype correlation which is crucial for screening and managing patients. Evaluation involves a combination of structural (CT/MRI) and functional imaging. MIBG remains helpful for PC assessment but novel PET ligands (18F-DOPA, 68Ga-DOTATATE, 18F-FDG) aid in the detection of extra-adrenal paragangliomas, recurrence, and metastatic disease. The treatment paradigm has shifted toward personalized medicine, incorporating genetic insights to tailor interventions, particularly surgical approaches and novel therapeutics such as radiolabeling of somatostatin analogs with lutetium and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Matti L Gild
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia.
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia.
| | - Kimchi Do
- Department of Endocrinology, Nepean Hospital, Sydney, Australia
| | - Venessa H M Tsang
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Lyndal J Tacon
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Roderick J Clifton-Bligh
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Bruce G Robinson
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| |
Collapse
|
9
|
Freitas-Castro F, Almeida MQ. Personalized management for phaeochromocytomas and paragangliomas in Latin America: A genetic perspective. Best Pract Res Clin Endocrinol Metab 2025; 39:101922. [PMID: 39244493 DOI: 10.1016/j.beem.2024.101922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors with clinical heterogeneity and a high association with hereditary disease, affecting approximately 30 % of the cases. Differences in the presentation and genetic etiologies of PPGLs have been demonstrated between Chinese and European patients. The frequency of germline genetic diagnosis was remarkably higher in Brazilian patients (∼50 %) compared with other cohorts (Chinese 21 %, European 31 %, and The Cancer Genome Atlas Program cohort 27 %). Interestingly, germline SDHB genetic defects were also more prevalent in Brazilian patients (17 %) with PPGLs when compared with other cohorts (3-9 %). The SDHB exon 1 deletion was responsible for approximately 50 % of the SDHB pathogenic/likely pathogenic variants in Brazilian patients with PPGLs due to a founder effect. The germline SDHB exon 1 deletion represents ∼10 % of the germline drivers in Brazilian patients (and possibly in Latin America). Therefore, a single diagnostic PCR for the SDHB exon 1 deletion might be very useful in clinical practice for genetic testing and counseling of patients with PPGLs in Latin America.
Collapse
Affiliation(s)
- Felipe Freitas-Castro
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Madson Q Almeida
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil; Unidade de Oncologia Endócrina, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil.
| |
Collapse
|
10
|
de Bresser CJM, de Krijger RR. The Molecular Classification of Pheochromocytomas and Paragangliomas: Discovering the Genomic and Immune Landscape of Metastatic Disease. Endocr Pathol 2024; 35:279-292. [PMID: 39466488 DOI: 10.1007/s12022-024-09830-3] [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] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs, together PPGLs) are the most hereditary tumors known. PPGLs were considered benign, but the fourth edition of the World Health Organisation (WHO) classification redefined all PPGLs as malignant neoplasms with variable metastatic potential. The metastatic rate differs based on histopathology, genetic background, size, and location of the tumor. The challenge in predicting metastatic disease lies in the absence of a clear genotype-phenotype correlation among the more than 20 identified genetic driver variants. Recent advances in molecular clustering based on underlying genetic alterations have paved the way for improved cluster-specific personalized treatments. However, despite some clusters demonstrating a higher propensity for metastatic disease, cluster-specific therapies have not yet been widely adopted in clinical practice. Comprehensive genomic profiling and transcriptomic analyses of large PPGL cohorts have identified potential new biomarkers that may influence metastatic potential. It appears that no single biomarker alone can reliably predict metastatic risk; instead, a combination of these biomarkers may be necessary to develop an effective prediction model for metastatic disease. This review evaluates current guidelines and recent genomic and transcriptomic findings, with the aim of accurately identifying novel biomarkers that could contribute to a predictive model for mPPGLs, thereby enhancing patient care and outcomes.
Collapse
Affiliation(s)
- Carolijn J M de Bresser
- Department of Vascular Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| |
Collapse
|
11
|
Jiang J, Liu Y. Recent discoveries of Sino-Caucasian differences in the genetics of phaeochromocytomas and paragangliomas. Best Pract Res Clin Endocrinol Metab 2024; 38:101928. [PMID: 39191630 DOI: 10.1016/j.beem.2024.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) represent the highest degree of heritability of any known tumor types in humans. Previous studies have characterized a dramatic difference between Chinese and European Caucasians with regards to both genetics and clinical features of PPGLs. The proportion of PGLs in Chinese patients was higher than in Caucasians, and the prevalence of metastasis was much lower in Chinese patients. Compared with Caucasians, there were more pathogenic variants (PVs) found in HRAS and FGFR1, but less in NF1 and SDHB. There were less germline PVs found in Chinese patients. Importantly, in Chinese patients, there was a large proportion of PGLs with PVs found in HRAS and FGFR1, mostly with epinephrine-producing capacity. This finding provided solid evidence that genetics (cluster 1 vs. 2), rather than location (PCC vs. PGL), determines the catecholamine-producing phenotype. Besides, the lower prevalence of SDHB partially explained lower occurrence of metastatic lesions in Chinese patients. These findings underscore the importance of considering ethnic differences when evaluating PPGLs and patient outcomes.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, China.
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
12
|
Casey RT, Hendriks E, Deal C, Waguespack SG, Wiegering V, Redlich A, Akker S, Prasad R, Fassnacht M, Clifton-Bligh R, Amar L, Bornstein S, Canu L, Charmandari E, Chrisoulidou A, Freixes MC, de Krijger R, de Sanctis L, Fojo A, Ghia AJ, Huebner A, Kosmoliaptsis V, Kuhlen M, Raffaelli M, Lussey-Lepoutre C, Marks SD, Nilubol N, Parasiliti-Caprino M, Timmers HHJLM, Zietlow AL, Robledo M, Gimenez-Roqueplo AP, Grossman AB, Taïeb D, Maher ER, Lenders JWM, Eisenhofer G, Jimenez C, Pacak K, Pamporaki C. International consensus statement on the diagnosis and management of phaeochromocytoma and paraganglioma in children and adolescents. Nat Rev Endocrinol 2024; 20:729-748. [PMID: 39147856 DOI: 10.1038/s41574-024-01024-5] [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: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Phaeochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumours that arise not only in adulthood but also in childhood and adolescence. Up to 70-80% of childhood PPGL are hereditary, accounting for a higher incidence of metastatic and/or multifocal PPGL in paediatric patients than in adult patients. Key differences in the tumour biology and management, together with rare disease incidence and therapeutic challenges in paediatric compared with adult patients, mandate close expert cross-disciplinary teamwork. Teams should ideally include adult and paediatric endocrinologists, oncologists, cardiologists, surgeons, geneticists, pathologists, radiologists, clinical psychologists and nuclear medicine physicians. Provision of an international Consensus Statement should improve care and outcomes for children and adolescents with these tumours.
Collapse
Affiliation(s)
- Ruth T Casey
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
- Department of Endocrinology, Cambridge Cancer Centre and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Emile Hendriks
- Department of Paediatric Diabetes and Endocrinology, Cambridge Cancer Centre and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Cheri Deal
- Endocrine and Diabetes Service, CHU Sainte-Justine and University of Montreal, Montreal, Québec, Canada
| | - Steven G Waguespack
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Verena Wiegering
- University Children's Hospital, Department of Paediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany
| | - Antje Redlich
- Paediatric Oncology Department, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Scott Akker
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Rathi Prasad
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Roderick Clifton-Bligh
- Department of Diabetes and Endocrinology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Laurence Amar
- Université de Paris, Paris, France
- Hypertension Unit, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Stefan Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Paediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Athens, Greece
| | | | - Maria Currás Freixes
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Ronald de Krijger
- Princess Maxima Center for Paediatric Oncology, Utrecht, Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Luisa de Sanctis
- Department of Public Health and Paediatric Sciences, University of Turin, Turin, Italy
| | - Antonio Fojo
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Amol J Ghia
- Department of Radiation Oncology, University Hospital of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Angela Huebner
- Department of Paediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, UK
- Blood and Transplant Research Unit in Organ Donation and Transplantation, National Institute for Health Research, University of Cambridge, Cambridge, UK
| | - Michaela Kuhlen
- Paediatrics and Adolescent Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Marco Raffaelli
- U.O.C. Chirurgia Endocrina e Metabolica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Charlotte Lussey-Lepoutre
- Service de médecine nucléaire, Inserm U970, Sorbonne université, Groupe hospitalier Pitié-Salpétrière, Paris, France
| | - Stephen D Marks
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust and NIHR GOSH Biomedical Research Centre, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mirko Parasiliti-Caprino
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti, Turin, Italy
| | - Henri H J L M Timmers
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Anna Lena Zietlow
- Clinical Child and Adolescent Psychology, Institute of Clinical Psychology and Psychotherapy, Department of Psychology, TU Dresden, Dresden, Germany
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, PARCC, INSERM, Paris, France
- Service de Génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Ashley B Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Graeme Eisenhofer
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD, USA
| | - Christina Pamporaki
- Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
13
|
Martínez de Lapiscina I, Diego E, Baquero C, Fernández E, Menendez E, Moure MD, Ruiz de Azua T, Castaño L, Valdés N, on behalf of the Collaborative Working Group. Novel Gene Variants in a Nationwide Cohort of Patients with Pheochromocytoma and Paraganglioma. Int J Mol Sci 2024; 25:12056. [PMID: 39596125 PMCID: PMC11593415 DOI: 10.3390/ijms252212056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs), denoted PPGLs, are rare neuroendocrine tumours and are highly heterogeneous. The phenotype-genotype correlation is poor; therefore, additional studies are needed to understand their pathogenesis. We describe the clinical characteristics of 63 patients with PPGLs and perform a genetic study. Genetic screening was performed via a targeted gene panel, and clinical variables were compared among patients with a positive molecular diagnosis and negative ones in both PCC and PGL cohorts. The mean age of patients with PCC was 50.0, and the mean age of those with PGL was 54.0. Disease-causing germline variants were identified in 16 individuals (25.4%), twelve and five patients with PCC and PGL, respectively. Genetically positive patients were younger at diagnosis in both cohorts. Variants in genes associated with either isolated PPGLs or syndromic forms of the disease were detected in a cohort of PPGLs. We have identified novel variants in known genes and set the importance of genetic screening to every patient with PPGLs, with a special focus on the young. A longer follow up of patients with variants in genes associated with syndromic forms is of clinical value.
Collapse
Affiliation(s)
- Idoia Martínez de Lapiscina
- Biobizkaia Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), European Reference Network on Rare Endocrine Conditions (Endo-ERN), Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | - Estrella Diego
- Biobizkaia Health Research Institute, Endocrinology and Nutrition Department, Cruces University Hospital, Plaza de Cruces s/n, 48903 Barakaldo, Spain; (E.D.); (E.F.)
| | - Candela Baquero
- Biobizkaia Health Research Institute, University of the Basque Country (UPV-EHU), Plaza de Cruces s/n, 48903 Barakaldo, Spain;
| | - Elsa Fernández
- Biobizkaia Health Research Institute, Endocrinology and Nutrition Department, Cruces University Hospital, Plaza de Cruces s/n, 48903 Barakaldo, Spain; (E.D.); (E.F.)
| | - Edelmiro Menendez
- Principality of Asturias Health Research Institute, Department of Endocrinology and Nutrition, Asturias Central University Hospital, Oviedo University, CIBERER. Av. Roma s/n, 33011 Oviedo, Spain;
| | - Maria Dolores Moure
- Biobizkaia Health Research Institute, Endocrinology and Nutrition Department, Cruces University Hospital, UPV-EHU, Plaza de Cruces s/n, 48903 Barakaldo, Spain;
| | - Teresa Ruiz de Azua
- Endocrinology Department, Urduliz Hospital, Goieta 32, 48610 Urduliz, Spain;
| | - Luis Castaño
- Biobizkaia Health Research Institute, Pediatric Endocrinology Department, Cruces University Hospital, UPV-EHU, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Endo-ERN, Plaza de Cruces s/n, 48903 Barakaldo, Spain;
| | - Nuria Valdés
- Biobizkaia Health Research Institute, Endocrinology and Nutrition Department, Cruces University Hospital, UPV-EHU, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CI-BERDEM), Endo-ERN, Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | | |
Collapse
|
14
|
Nazari MA, Jha A, Kuo MJM, Patel M, Prodanov T, Rosenblum JS, Talvacchio S, Derkyi A, Charles K, Pacak K. Paediatric phaeochromocytoma and paraganglioma: A clinical update. Clin Endocrinol (Oxf) 2024; 101:446-454. [PMID: 37515400 DOI: 10.1111/cen.14955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Paediatric phaeochromocytomas and paragangliomas (PPGLs), though rare tumours, are associated with significant disability and death in the most vulnerable of patients early in their lives. However, unlike cryptogenic and insidious disease states, the clinical presentation of paediatric patients with PPGLs can be rather overt, allowing early diagnosis, granted that salient findings are recognized. Additionally, with prompt and effective intervention, prognosis is favourable if timely intervention is implemented. For this reason, this review focuses on four exemplary paediatric cases, succinctly emphasizing the now state-of-the-art concepts in paediatric PPGL management.
Collapse
Affiliation(s)
- Matthew A Nazari
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhishek Jha
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Mickey J M Kuo
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mayank Patel
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Tamara Prodanov
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Jared S Rosenblum
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Talvacchio
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Alberta Derkyi
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Kailah Charles
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Karel Pacak
- Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Jo KH, Lee J, Yoo J, Kim HS, Kim ES, Han JH, Jang YS, Yun JS, Son JW, Yoo SJ, Lee SH, Lim DJ, Kwon HS, Lee S, Moon S, Kim M. Germline Mutations and Phenotypic Associations in Korean Patients With Pheochromocytoma and Paraganglioma: A Multicenter Study and Literature Review. Ann Lab Med 2024; 44:591-597. [PMID: 39069753 PMCID: PMC11375186 DOI: 10.3343/alm.2023.0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/17/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
Genetic testing is recommended for all patients with pheochromocytomas and paragangliomas (PPGL) to establish genotype-phenotype associations. We investigated germline mutations in 59 patients with PPGL at six Korean university hospitals using next-generation sequencing (NGS) targeting 38 PPGL-associated genes, including those recommended by the Korean PPGL Task Force. Germline mutations were identified in 13 patients (22%), and affected four genes: RET, NF1, VHL, and SDHD. Germline mutations were significantly associated with a family history of PPGL, smaller tumor size, and the presence of other types of tumors. Using 95 Korean PPGL cases with germline mutations identified through a literature review and 13 cases from our cohort, we characterized genotype-phenotype correlations. Mutation hotspots were identified in specific codons of RET (codons 631 and 634), VHL (157 and 167), and SDHB (131 and 253). NF1 mutations varied, indicating the absence of common hotspots. These findings highlight the efficacy of the recommended NGS panel for Korean patients with PPGL and the importance of genetic testing in establishing clinical management and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Kwan Hoon Jo
- Division of Endocrinology & Metabolism, Department of Internal Medicine, Incheon St. Mary’s hospital, The Catholic University of Korea, Seoul, Korea
| | - Jaewoong Lee
- Department of Laboratory Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jaeeun Yoo
- Department of Laboratory Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hoon Seok Kim
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Sook Kim
- Division of Endocrinology & Metabolism, Department of Internal Medicine, Incheon St. Mary’s hospital, The Catholic University of Korea, Seoul, Korea
| | - Je Ho Han
- Division of Endocrinology & Metabolism, Department of Internal Medicine, Incheon St. Mary’s hospital, The Catholic University of Korea, Seoul, Korea
| | - Yi Sun Jang
- Department of Internal Medicine, Daejeon St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Seung Yun
- Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jang Won Son
- Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soon Jib Yoo
- Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Hwan Lee
- Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong Jun Lim
- Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyuk-Sang Kwon
- Department of Internal Medicine, Yeouido St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seungok Lee
- Department of Laboratory Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sungdae Moon
- Division of Endocrinology & Metabolism, Department of Internal Medicine, Incheon St. Mary’s hospital, The Catholic University of Korea, Seoul, Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
16
|
Cortez BN, Kuo MJM, Jha A, Patel M, Carrasquillo JA, Prodanov T, Charles KM, Talvacchio S, Derkyi A, Lin FI, Taïeb D, Del Rivero J, Pacak K. Case Series: ATRX Variants in Four Patients with Metastatic Pheochromocytoma. Front Endocrinol (Lausanne) 2024; 15:1399847. [PMID: 39351526 PMCID: PMC11439680 DOI: 10.3389/fendo.2024.1399847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 10/04/2024] Open
Abstract
Few reports have highlighted the rare presence of somatic ATRX variants in clinically aggressive, metastatic pheochromocytoma/paraganglioma (PCC/PGL); however, none have addressed detailed clinical presentation (including biochemistry and imaging) and management of these patients. Here, we address these clinical features and management based on four PCC patients with somatic ATRX variants from our National Institutes of Health PCC/PGL cohort. A total of 192 patients underwent exome sequencing (germline, somatic, or both), and four males were found to have somatic ATRX variants (with additional somatic VHL and FH oncogenic variants in patients 2 and 4, respectively). Per-lesion and per-patient comparisons were performed among functional imaging scans performed at the NIH. Biochemical phenotype and response to systemic treatment were evaluated. This mini-series supports prior studies showing aggressive/metastatic PCC in patients with somatic ATRX variants, as all developed widespread metastatic disease. All four PCC patients presented with noradrenergic biochemical phenotype, and some with significant elevation in 3-methoxytyramine. 18F-FDOPA PET/CT was found to be the superior functional imaging modality, with 100% lesion detection rate when compared to that of 68Ga-DOTATATE, 18F-FDG, 18F-FDA, and 123I-MIBG scans. While patients did not respond to chemotherapy or tyrosine kinase inhibitors, they responded to targeted radiotherapy using high-specific-activity 131I-MIBG (Azedra®) or 177Lu-DOTATATE (Lutathera®).
Collapse
Affiliation(s)
- Briana N. Cortez
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mickey J. M. Kuo
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mayank Patel
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- Center for Cancer Research, Laboratory of Pathology, National Cancer Institute, Bethesda, MD, United States
| | - Jorge A. Carrasquillo
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tamara Prodanov
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Kailah M. Charles
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Sara Talvacchio
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Alberta Derkyi
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Frank I. Lin
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital & Centre de Recherches en Cancérologie de Marseille (CERIMED) & French Institute of Health and Medical Research (Inserm) UMR1068 Marseille Cancerology Research Center, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Jaydira Del Rivero
- Developemental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
17
|
Cascón A, Robledo M. Clinical and molecular markers guide the genetics of pheochromocytoma and paraganglioma. Biochim Biophys Acta Rev Cancer 2024; 1879:189141. [PMID: 38908536 DOI: 10.1016/j.bbcan.2024.189141] [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: 05/07/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Over the past two decades, research into the genetic susceptibility behind pheochromocytoma and paraganglioma (PPGL) has surged, ranking them among the most heritable tumors. Massive sequencing combined with careful patient selection has so far identified more than twenty susceptibility genes, leading to an over-detection of variants of unknown significance (VUS) that require precise molecular markers to determine their pathogenic role. Moreover, some PPGL patients remain undiagnosed, possibly due to mutations in regulatory regions of already known genes or mutations in undiscovered genes. Accurate classification of VUS and identification of new genes require well-defined clinical and molecular markers that allow effective genetic diagnosis of most PPGLs.
Collapse
Affiliation(s)
- Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
18
|
Tung N, Ricker C, Messersmith H, Balmaña J, Domchek S, Stoffel EM, Almhanna K, Arun B, Chavarri-Guerra Y, Cohen SA, Cragun D, Crew KD, Hall MJ, Idos G, Lopez G, Pal T, Pirzadeh-Miller S, Pritchard C, Rana HQ, Swami U, Vidal GA. Selection of Germline Genetic Testing Panels in Patients With Cancer: ASCO Guideline. J Clin Oncol 2024; 42:2599-2615. [PMID: 38759122 DOI: 10.1200/jco.24.00662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 05/19/2024] Open
Abstract
PURPOSE To guide use of multigene panels for germline genetic testing for patients with cancer. METHODS An ASCO Expert Panel convened to develop recommendations on the basis of a systematic review of guidelines, consensus statements, and studies of germline and somatic genetic testing. RESULTS Fifty-two guidelines and consensus statements met eligibility criteria for the primary search; 14 studies were identified for Clinical Question 4. RECOMMENDATIONS Patients should have a family history taken and recorded that includes details of cancers in first- and second-degree relatives and the patient's ethnicity. When more than one gene is relevant based on personal and/or family history, multigene panel testing should be offered. When considering what genes to include in the panel, the minimal panel should include the more strongly recommended genes from Table 1 and may include those less strongly recommended. A broader panel may be ordered when the potential benefits are clearly identified, and the potential harms from uncertain results should be mitigated. Patients who meet criteria for germline genetic testing should be offered germline testing regardless of results from tumor testing. Patients who would not normally be offered germline genetic testing based on personal and/or family history criteria but who have a pathogenic or likely pathogenic variant identified by tumor testing in a gene listed in Table 2 under the outlined circumstances should be offered germline testing.Additional information is available at www.asco.org/molecular-testing-and-biomarkers-guidelines.
Collapse
Affiliation(s)
- Nadine Tung
- Beth Israel Deaconess Medical Center, Sharon, MA
| | | | | | | | | | | | | | - Banu Arun
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yanin Chavarri-Guerra
- Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Mexico City, Mexico
| | | | | | | | | | - Gregory Idos
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ghecemy Lopez
- USC Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Sara Pirzadeh-Miller
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | - Umang Swami
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| | - Gregory A Vidal
- The West Cancer Center and Research Institute and The University of Tennessee Health Sciences Center, Germantown, TN
| |
Collapse
|
19
|
Tarling JA, Kumar R, Ward LJ, Boot C, Wassif WS. Phaeochromocytoma and paraganglioma. J Clin Pathol 2024; 77:507-516. [PMID: 38453430 DOI: 10.1136/jcp-2023-209234] [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: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 03/09/2024]
Abstract
Phaeochromocytomas and paragangliomas are rare catecholamine-producing neuroendocrine tumours which can potentially cause catastrophic crises with high morbidity and mortality. This best practice article considers the causes and presentation of such tumours, screening and diagnostic tests, management of these patients and consideration of family members at risk.
Collapse
Affiliation(s)
- Julie Ann Tarling
- Clinical Biochemistry, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| | - Rajeev Kumar
- Diabetes and Endocrinology, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| | - Louise J Ward
- Clinical Biochemistry, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| | - Christopher Boot
- Blood Sciences, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - W S Wassif
- Clinical Biochemistry, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| |
Collapse
|
20
|
Cioppi F, Cantini G, Ercolino T, Chetta M, Zanatta L, Nesi G, Mannelli M, Maggi M, Canu L, Luconi M. Targeted Next Generation Sequencing molecular profiling and its clinical application in adrenocortical cancer. Eur J Endocrinol 2024; 191:17-30. [PMID: 38917236 DOI: 10.1093/ejendo/lvae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
OBJECTIVE Adrenal cortical carcinoma (ACC) is a rare malignancy with a generally poor but heterogeneous prognosis, especially depending on the tumour stage at diagnosis. Identification of somatic gene alterations combined with clinical/histopathological evaluation of the tumour can help improve prognostication. We applied a simplified targeted-Next-Generation Sequencing (NGS) panel to characterise the mutational profiles of ACCs, providing potentially relevant information for better patient management. DESIGN AND METHODS Thirty frozen tumour specimens from a local ACC series were retrospectively analysed by a custom-NGS panel (CDKN2A, CTNNB1, DAXX, MED12, NF1, PRKAR1A, RB1, TERT, TP53, ZNRF3) to detect somatic prioritised single-nucleotide variants. This cohort was integrated with 86 patients from the ACC-TCGA series bearing point-mutations in the same genes and their combinations identified by our panel. Primary endpoints of the analysis on the total cohort (113 patients) were overall survival (OS) and progression-free survival (PFS), and hazard ratio (HR) for the different alterations grouped by the signalling pathways/combinations affected. RESULTS Different PFS, OS, and HR were associated to the different pathways/combinations, being NF1 + TP53 and Wnt/β-catenin + Rb/p53 combined mutations the most deleterious, with a statistical significance for progression HR which is retained only in low-(I/II) stages-NF1 + TP53 combination: HR = 2.96[1.01-8.69] and HR = 13.23[3.15-55.61], all and low stages, respectively; Wnt/β-catenin + Rb/p53 combined pathways: HR = 6.47[2.54-16.49] and HR = 16.24[3.87-68.00], all and low-stages, respectively. CONCLUSIONS A simplified targeted-NGS approach seems the best routinely applicable first step towards somatic genetic characterisation of ACC for prognostic assessment. This approach proved to be particularly promising in low-stage cases, suggesting the need for more stringent surveillance and personalised treatment.
Collapse
Affiliation(s)
- Francesca Cioppi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
| | - Giulia Cantini
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| | - Tonino Ercolino
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Massimiliano Chetta
- Medical Genetics, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Cardarelli, Padiglione, 80131 Naples, Italy
| | - Lorenzo Zanatta
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Massimo Mannelli
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| | - Mario Maggi
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Letizia Canu
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Michaela Luconi
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| |
Collapse
|
21
|
Li Q, Lan Z, Jiang Y, Wang R, Li Z, Jiang X. Validation and Evaluation of 5 Scoring Systems for Predicting Metastatic Risk in Pheochromocytoma and Paraganglioma. Am J Surg Pathol 2024; 48:855-865. [PMID: 38712603 DOI: 10.1097/pas.0000000000002238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Currently, 5 scoring systems have been proposed in the literature for predicting metastatic risk in pheochromocytoma and paraganglioma (PPGL): Pheochromocytoma of the Adrenal Gland Scaled Score (PASS), Grading System for Adrenal Pheochromocytoma and Paraganglioma (GAPP), Composite Pheochromocytoma/paraganglioma Prognostic Score (COPPS), Age, Size, Extra-adrenal location, Secretion type (ASES) score, and Size, Genetic, Age, and PASS (SGAP) model. To validate and evaluate these 5 scoring systems, we conducted a retrospective review of cases diagnosed as PPGL at the Department of Pathology, West China Hospital of Sichuan University, between January 2012 and December 2019. A total of 185 PPGL cases were included, comprising 35 cases with metastasis and 150 cases remained metastasis-free for over 8 years after surgery. The criteria of the 5 scoring systems were used for scoring and risk classification. The predictive performance of the 5 scoring systems was validated, compared, and evaluated using concordance index (C-index) and decision curve analysis (DCA). The C-indices for PASS, GAPP, and SGAP were 0.600, 0.547, and 0.547, respectively, indicating low discriminative ability. In contrast, COPPS and ASES had C-indices of 0.740 and 0.706, respectively, indicating better discriminative performance. DCA also showed that the predictive capability of COPPS was superior to that of ASES, with both outperformed PASS, while PASS had better predictive ability than GAPP and SGAP. Our analysis indicated that pathology-based scoring systems cannot accurately predict metastatic risk of PPGL. Establishing a precise prediction system requires integrating clinical, pathologic, and molecular information, using a scientific methodology for predictive factor selection and weight assessment.
Collapse
Affiliation(s)
- Qin Li
- Departments of Pathology
- Neurosurgery, West China Hospital of Sichuan University
| | - Zhigang Lan
- Department of Pathology, Chengdu Fifth People's Hospital, The Fifth People's Hospital Affiliated to Chengdu University of Traditional Chinese Medicine
| | | | | | | | - Xiaolin Jiang
- Department of Pathology, Guangyuan Central Hospital, Chengdu, Sichuan Province, China
| |
Collapse
|
22
|
Ameti A, Kopp PA, Pitteloud N, Wuerzner G, Grouzmann E, Matter M, Lamine F, Phan O. Hypertension in a Patient With Polycystic Kidney Disease Complicated by Concomitant Pheochromocytoma. AACE Clin Case Rep 2024; 10:136-139. [PMID: 39100633 PMCID: PMC11294747 DOI: 10.1016/j.aace.2024.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 08/06/2024] Open
Abstract
Background Due to the high prevalence of hypertension in patients with autosomal dominant polycystic kidney disease (ADPKD) and advanced chronic kidney disease, diagnosing secondary hypertension poses challenges. We present a rare case of pheochromocytoma in an ADPKD patient to highlight the diagnostic difficulties in identifying secondary hypertension due to pheochromocytoma/paraganglioma (PPGL) in end-stage renal disease (ESRD) patients. Case Report A 48-year-old female with ADPKD and ESRD experienced recurrent hypertensive crises (up to 220/135 mmHg) accompanied by palpitations and tremors that recurred over the past 2 years. Introduction of a betablocker to the antihypertensive therapy aggravated her symptoms. The initial documentation of elevated urinary metanephrines was interpreted as false positive finding due to renal failure. Subsequent measurements of free plasma metanephrines revealed significant elevations raising suspicion of PPGL. Magnetic resonance imaging identified a 29 mm right adrenal mass. The patient underwent right adrenalectomy resulting in resolution of the hypertensive crises. Discussion The diagnosis of PPGLs can present significant challenges and is further complicated in ESRD due to nonspecific clinical symptoms and diagnostic pitfalls. Less than 20 PPGL cases have been reported in patients with ESRD. The intolerance of beta-blocker therapy, as well as the use of a scoring system for the likelihood of PPGL should have raised suspicion. Conclusion PPGL should be considered in all patients with uncontrolled hypertension and beta-blockers intolerance, even in the presence of other etiologic mechanisms such as ESRD. Measuring free plasma metanephrines provides the most reliable biochemical screening in the context of impaired renal function.
Collapse
Affiliation(s)
- Adelina Ameti
- Division of Endocrinology, Diabetes & Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Peter A Kopp
- Division of Endocrinology, Diabetes & Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Nelly Pitteloud
- Division of Endocrinology, Diabetes & Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Grégoire Wuerzner
- Division of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Eric Grouzmann
- Division of Clinical Pharmacology, Catecholamine and Peptides Laboratory, CHUV, Lausanne, Switzerland
| | - Maurice Matter
- Division of Visceral Surgery, University Hospital CHUV, Lausanne, Switzerland
| | - Faiza Lamine
- Division of Endocrinology, Diabetes & Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Olivier Phan
- Division of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
23
|
de Miguel VC, Aparicio LS, Sansó G, Paissan AL, Lupi SN, Belli SH, Tkatch J, Marín MJ, Barontini MB. Seventy years of pheochromocytomas and paragangliomas in Argentina. The FRENAR database. HIPERTENSION Y RIESGO VASCULAR 2024; 41:170-178. [PMID: 38693013 DOI: 10.1016/j.hipert.2024.04.001] [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: 01/04/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 05/03/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are neuroendocrine tumors characterized by the excessive production of catecholamines. This study aims to describe the clinical characteristics of PPGL cases in Argentina over recent decades. A multicenter retrospective cross-sectional analysis was carried out using a database comprising both pediatric and adult patients with confirmed PPGL diagnoses based on pathological reports. A cohort of 486 patients with PPGL was recruited. Women represent 58.4% of the patients, with a mean age of 38.3 years old at the time of diagnosis and 15.2% of the patients were under the age of 18. Hypertension, as well as classic signs and symptoms, were present in 80.9% of the patients. The adrenal incidentaloma, as a mode of presentation, increased in the last two decades rising from 3.9% (1953-2000) to 21.8% (2001-2022), p<0.001. Most tumors were located within the adrenal glands, accounting 83.0% of the cases, with bilateral occurrences noted in 20.0%. The median tumor size was 4.8cm. Local recurrence and metastases were observed in 10.9% and 12.2%. Out of 412 patients, 87.0% exhibited urinary excretion elevation of catecholamines and/or their metabolites. Furthermore, 148 patients, representing 30.4% of the study population, displayed a distinct genetic profile indicative of hereditary syndromes. The distribution of hereditary syndromes revealed that MEN2, VHL, and PGL4 constituted the most prevalent syndromes. This population-based study, spanning seven decades, offers valuable insights into the demographic and clinical characteristics of PPGL patients in Argentina.
Collapse
Affiliation(s)
| | | | - G Sansó
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - A L Paissan
- Hospital Italiano de Buenos Aires, Argentina
| | - S N Lupi
- Hospital Ramos Mejía, Buenos Aires, Argentina
| | - S H Belli
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | - J Tkatch
- Hospital Durand, Buenos Aires, Argentina
| | - M J Marín
- Hospital Italiano de Buenos Aires, Argentina
| | - M B Barontini
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
24
|
Cardot-Bauters C, Vantyghem MC, Do Cao C, Desailloud R, Joubert M, Coppin L, Odou MF, Pigny P. Genetic predisposition to pheochromocytoma and paraganglioma: 21 years of experience in the field. ANNALES D'ENDOCRINOLOGIE 2024; 85:276-283. [PMID: 38815921 DOI: 10.1016/j.ando.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/20/2024] [Accepted: 05/04/2024] [Indexed: 06/01/2024]
Abstract
CONTEXT Pheochromocytoma and paraganglioma (PPGL) are rare neuroendocrine tumors with high heritability, justifying systematic genetic screening for a germline variant in one of the twenty predisposing genes described to date. PURPOSE To describe the experience of one endocrine oncogenetic laboratory over a period of 21 years (2001-2022), from the beginning of PPGL genotyping with Sanger sequencing in 2001 to the implementation of next-generation sequencing (NGS). METHOD The activity database of an academic oncogenetic laboratory was searched to extract patients/relatives identified with a pathogenic variant/likely pathogenic variant (PV/LPV) over a period of 21 years. Clinical and genetic data were compared. RESULTS In total, 606 index cases with PPGL and 444 relatives were genotyped. Genotyping of index cases was performed by Sanger sequencing and gene deletion analysis in 327 cases and by NGS in 279. Germline PV/LPV spanning 10 genes was identified in 165 index cases (27.2%). Several recurrent PV/LPVs in SDHx were observed in non-related index cases, the most frequent being SDHD, c.170-1G>T (n=28). This subgroup showed great phenotypic variability both between and within families in terms of both tumor location and number. Four patients (1.1%) with PV/LPV in SDHx had 3PA (Pituitary Adenoma and pheochromocytoma/paraganglioma) syndrome. 258 relatives (58.1%) had inherited a PV/LPV in one driver gene. The rate of PV/LPV carriers who were symptomatic at first imaging evaluation was 32%, but varied between<20% in SDHB and SDHC and >50% in SDHD, VHL and MAX. CONCLUSION Our experience confirmed previously established genotype-phenotype correlations, but also highlights atypical clinical presentations, even for the same genetic variant. These data must be taken into account for optimal patient follow-up and management.
Collapse
Affiliation(s)
- Catherine Cardot-Bauters
- Service d'endocrinologie, diabétologie-métabolisme, hôpital Claude-Huriez, CHU, 59037 Lille cedex, France
| | - Marie-Christine Vantyghem
- Service d'endocrinologie, diabétologie-métabolisme, hôpital Claude-Huriez, CHU, 59037 Lille cedex, France
| | - Christine Do Cao
- Service d'endocrinologie, diabétologie-métabolisme, hôpital Claude-Huriez, CHU, 59037 Lille cedex, France
| | - Rachel Desailloud
- Service d'endocrinologie-diabétologie, nutrition, hôpital Sud nord, CHU, 80054 Amiens cedex 1, France
| | - Michael Joubert
- Service d'endocrinologie, CHU Côte de Nacre, 14000 Caen cedex, France
| | - Lucie Coppin
- Laboratoire de biochimie « hormonologie-métabolisme-nutrition-oncologie », UF oncogénétique moléculaire, laboratoire de biologie médicale de référence, CHU de Lille, 59037 Lille cedex, France; Inserm UMR 1277, CANTHER, université de Lille, 59045 Lille cedex, France
| | - Marie-Francoise Odou
- Laboratoire de biochimie « hormonologie-métabolisme-nutrition-oncologie », UF oncogénétique moléculaire, laboratoire de biologie médicale de référence, CHU de Lille, 59037 Lille cedex, France; Inserm, CHU Lille, U1286 - Infinite, université de Lille, 59045 Lille cedex, France
| | - Pascal Pigny
- Laboratoire de biochimie « hormonologie-métabolisme-nutrition-oncologie », UF oncogénétique moléculaire, laboratoire de biologie médicale de référence, CHU de Lille, 59037 Lille cedex, France; Inserm UMR 1277, CANTHER, université de Lille, 59045 Lille cedex, France.
| |
Collapse
|
25
|
Iguchi DYV, Martins Filho SN, Soares IC, Siqueira SAC, Alves VAF, Assato AK, Yang JH, Almeida MQ, Villares Fragoso MCB, Fagundes GFC, Mendonca BB, Lourenço Junior DM, Hoff AO, Castroneves LA, Ferraz-de-Souza B, Giannella MLCC, Pereira MAA. Identification of Predictors of Metastatic Potential in Paragangliomas to Develop a Prognostic Score (PSPGL). J Endocr Soc 2024; 8:bvae093. [PMID: 38799767 PMCID: PMC11112433 DOI: 10.1210/jendso/bvae093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Indexed: 05/29/2024] Open
Abstract
Context Paragangliomas (PGLs) are rare tumors in adrenal and extra-adrenal locations. Metastasis are found in approximately 5% to 35% of PGLs, and there are no reliable predictors of metastatic disease. Objective This work aimed to develop a prognostic score of metastatic potential in PGLs. Methods A retrospective analysis was conducted of clinical data from a cohort with PGLs and tumor histological assessment. Patients were divided into metastatic PGL (presence of metastasis) and nonmetastatic PGL (absence of metastasis ≥96 months of follow-up) groups. Univariate and multivariable analysis were performed to identify predictors of metastatic potential. A prognostic score was developed based on coefficients of multivariable analysis. Kaplan-Meier curves were generated to estimate disease-specific survival (DSS). Results Out of 263 patients, 35 patients had metastatic PGL and 110 patients had nonmetastatic PGL. In multivariable analysis, 4 features were independently related to metastatic disease and composed the Prognostic Score of Paragangliomas (PSPGL): presence of central or confluent necrosis (33 points), more than 3 mitosis/10 high-power field (HPF) (28 points), extension into adipose tissue (20 points), and extra-adrenal location (19 points). A PSPGL of 24 or greater showed similar sensitivity with higher specificity than the Pheochromocytoma of the Adrenal Gland Scaled Score (PASS) and Grading System for Adrenal Pheochromocytoma and Paraganglioma (GAPP). PSPGL less than or equal to 20 was associated with a risk of metastasis of approximately 10%, whereas a PSPGL of 40 or greater was associated with approximately 80%. The presence of metastasis and Ki-67 of 3% or greater were related to lower DSS. Conclusion The PSPGL, composed of 4 easy-to-assess parameters, demonstrated good performance in predicting metastatic potential and good ability in estimating metastasis risk.
Collapse
Affiliation(s)
- Daniela Yone Veiga Iguchi
- Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | | | - Iberê Cauduro Soares
- Divisão de Anatomia Patológica, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Sheila Aparecida Coelho Siqueira
- Divisão de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Venâncio Avancini Ferreira Alves
- Laboratório de Investigaçãoc Médica LIM/14, Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Aline Kawassaki Assato
- Laboratório de Investigaçãoc Médica LIM/14, Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ji Hoon Yang
- Clínica de Endocrinologia, Hospital do Servidor Público Municipal de São Paulo, São Paulo 01532-000, Brazil
| | - Madson Q Almeida
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Maria Candida Barisson Villares Fragoso
- Laboratório de Hormônios e Genética Molecular LIM/42, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Gustavo Freitas Cardoso Fagundes
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Berenice B Mendonca
- Laboratório de Hormônios e Genética Molecular LIM/42, Laboratório de Sequenciamento em Larga Escala (SELA), Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Delmar Muniz Lourenço Junior
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ana O Hoff
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Luciana Audi Castroneves
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Bruno Ferraz-de-Souza
- Laboratório de Endocrinologia Celular e Molecular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo 01246-903, Brazil
- School of Medicine, University of Notre Dame Australia, Fremantle WA 6160, Australia
| | - Maria Lucia Cardillo Correa Giannella
- Laboratório de Carboidratos e Radioimunoensaio LIM/18, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Maria Adelaide Albergaria Pereira
- Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| |
Collapse
|
26
|
Walker TJ, Reyes-Alvarez E, Hyndman BD, Sugiyama MG, Oliveira LCB, Rekab AN, Crupi MJF, Cabral-Dias R, Guo Q, Dahia PLM, Richardson DS, Antonescu CN, Mulligan LM. Loss of tumor suppressor TMEM127 drives RET-mediated transformation through disrupted membrane dynamics. eLife 2024; 12:RP89100. [PMID: 38687678 PMCID: PMC11060712 DOI: 10.7554/elife.89100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Internalization from the cell membrane and endosomal trafficking of receptor tyrosine kinases (RTKs) are important regulators of signaling in normal cells that can frequently be disrupted in cancer. The adrenal tumor pheochromocytoma (PCC) can be caused by activating mutations of the rearranged during transfection (RET) receptor tyrosine kinase, or inactivation of TMEM127, a transmembrane tumor suppressor implicated in trafficking of endosomal cargos. However, the role of aberrant receptor trafficking in PCC is not well understood. Here, we show that loss of TMEM127 causes wildtype RET protein accumulation on the cell surface, where increased receptor density facilitates constitutive ligand-independent activity and downstream signaling, driving cell proliferation. Loss of TMEM127 altered normal cell membrane organization and recruitment and stabilization of membrane protein complexes, impaired assembly, and maturation of clathrin-coated pits, and reduced internalization and degradation of cell surface RET. In addition to RTKs, TMEM127 depletion also promoted surface accumulation of several other transmembrane proteins, suggesting it may cause global defects in surface protein activity and function. Together, our data identify TMEM127 as an important determinant of membrane organization including membrane protein diffusability and protein complex assembly and provide a novel paradigm for oncogenesis in PCC where altered membrane dynamics promotes cell surface accumulation and constitutive activity of growth factor receptors to drive aberrant signaling and promote transformation.
Collapse
Affiliation(s)
- Timothy J Walker
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Eduardo Reyes-Alvarez
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Brandy D Hyndman
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Michael G Sugiyama
- Department of Chemistry and Biology, Toronto Metropolitan UniversityTorontoCanada
| | - Larissa CB Oliveira
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Aisha N Rekab
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Mathieu JF Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| | - Rebecca Cabral-Dias
- Department of Chemistry and Biology, Toronto Metropolitan UniversityTorontoCanada
| | - Qianjin Guo
- Division of Hematology and Medical Oncology, University of Texas Health Science CenterSan AntonioUnited States
| | - Patricia LM Dahia
- Division of Hematology and Medical Oncology, University of Texas Health Science CenterSan AntonioUnited States
| | - Douglas S Richardson
- Department of Molecular and Cellular Biology, Harvard Center for Biological Imaging, Scientific Image Analysis Group, Harvard UniversityCambridgeUnited States
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan UniversityTorontoCanada
| | - Lois M Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s UniversityKingstonCanada
| |
Collapse
|
27
|
Walker TJ, Reyes-Alvarez E, Hyndman BD, Sugiyama MG, Oliveira LC, Rekab AN, Crupi MJ, Cabral-Dias R, Guo Q, Dahia PL, Richardson DS, Antonescu CN, Mulligan LM. Loss of Tumour Suppressor TMEM127 Drives RET-mediated Transformation Through Disrupted Membrane Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.28.546955. [PMID: 37425958 PMCID: PMC10327082 DOI: 10.1101/2023.06.28.546955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Internalization from the cell membrane and endosomal trafficking of receptor tyrosine kinases (RTK) are important regulators of signaling in normal cells that can frequently be disrupted in cancer. The adrenal tumour pheochromocytoma (PCC) can be caused by activating mutations of the RET receptor tyrosine kinase, or inactivation of TMEM127, a transmembrane tumour suppressor implicated in trafficking of endosomal cargos. However, the role of aberrant receptor trafficking in PCC is not well understood. Here, we show that loss of TMEM127 causes wildtype RET protein accumulation on the cell surface, where increased receptor density facilitates constitutive ligand-independent activity and downstream signaling, driving cell proliferation. Loss of TMEM127 altered normal cell membrane organization and recruitment and stabilization of membrane protein complexes, impaired assembly, and maturation of clathrin coated pits, and reduced internalization and degradation of cell surface RET. In addition to RTKs, TMEM127 depletion also promoted surface accumulation of several other transmembrane proteins, suggesting it may cause global defects in surface protein activity and function. Together, our data identify TMEM127 as an important determinant of membrane organization including membrane protein diffusability, and protein complex assembly and provide a novel paradigm for oncogenesis in PCC where altered membrane dynamics promotes cell surface accumulation and constitutive activity of growth factor receptors to drive aberrant signaling and promote transformation.
Collapse
Affiliation(s)
- Timothy J. Walker
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Eduardo Reyes-Alvarez
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Brandy D. Hyndman
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Michael G. Sugiyama
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Larissa C.B. Oliveira
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Aisha N. Rekab
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Mathieu J.F. Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| | - Rebecca Cabral-Dias
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Qianjin Guo
- Division of Hematology and Medical Oncology, University of Texas Health Science Center, San Antonio, Texas, 78229, United States
| | - Patricia L.M. Dahia
- Division of Hematology and Medical Oncology, University of Texas Health Science Center, San Antonio, Texas, 78229, United States
| | - Douglas S. Richardson
- Department of Molecular and Cellular Biology; Harvard Center for Biological Imaging; Scientific Image Analysis Group, Harvard University, Cambridge, MA, USA
| | - Costin N. Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Lois M. Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute, and Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
28
|
Park YG, Park I, Kim Y, Lee HS, Lee W, Yoon S, Lee JL. Outcomes of systemic treatment according to germline mutational status in patients with metastatic pheochromocytoma and paraganglioma. Clin Genitourin Cancer 2024; 22:413-419. [PMID: 38228412 DOI: 10.1016/j.clgc.2023.12.012] [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: 07/13/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
INTRODUCTION Metastatic disease affects approximately 15% to 17% of patients with pheochromocytomas and paragangliomas (PPGLs). Unfortunately, treatment options for metastatic PPGLs are limited and rely on small, nonrandomized clinical trials. The impact of germline mutation status on systemic treatment outcomes remains unclear. To address these gaps, we retrospectively evaluated treatment outcomes in patients with PPGL. PATIENTS AND METHODS Between December 2004 and December 2021, 33 patients were diagnosed with metastatic PPGLs and received systemic treatment at the Department of Oncology, Asan Medical Center, Seoul, South Korea. RESULTS The median age of the patients was 49. Germline mutations were revealed in nine patients (39.1%) out of 23 who underwent germline testing, with SDHB mutation being the most frequent in 5 patients. Cyclophosphamide, vincristine, and dacarbazine (CVD) chemotherapy was administered to 18 patients, with an objective response rate (ORR) of 22% and a disease control rate (DCR) of 67%. The median progression-free survival (PFS) was 7.9 and the median overall survival (OS) was 36.2 months. Sunitinib was given to 6 patients, which had an ORR of 33%, a DCR of 83%, and a median PFS of 14.6 months. Notably, patients with SDHB/SDHD mutation (4 patients and one patient, respectively) who received CVD treatment had a significantly better OS than those without (median OS 94.0 months vs. 13.7 months, P = .01). CONCLUSION Our study reveals that CVD and sunitinib are effective treatments for metastatic PPGLs. The results are consistent with previous studies and patients with SDHB and SDHD mutations may benefit most from CVD treatment.
Collapse
Affiliation(s)
- Young-Gyu Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yongjae Kim
- Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho-Su Lee
- Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woochang Lee
- Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
29
|
Giorgi RB, Aroucha PT, Favreto TM, Montero MF, Velloni JMF, Korkes I, Ferreira EN, Olivati C, Lima JV, Kater CE, Costa-Barbosa FA. Pheochromocytoma/Paraganglioma (PPGL): A Misdiagnosed Cause of Hypertension during Pregnancy. Case Rep Obstet Gynecol 2024; 2024:6655229. [PMID: 38572182 PMCID: PMC10990643 DOI: 10.1155/2024/6655229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/12/2024] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Hypertension (HT) during pregnancy is not an infrequent obstetric problem, reaching a prevalence of 5-10%. This condition is highly associated with both maternal and fetal complications if not precisely diagnosed and managed. Even though primary HT, obesity, and preeclampsia are the main causes of HT in this period, other less familiar conditions must be considered during the investigation. Pheochromocytoma and paraganglioma (PPGL) are chromaffin cell tumors that produce, store, and secrete catecholamines, leading to HT and other adrenergic manifestations. Recognition of PPGL is crucial since misdiagnosis and improper management can lead to high morbidity and mortality, particularly during pregnancy. We report on two cases of PPGL diagnosed during pregnancy with different managements. Case 1 is a 25-year-old female at 31 weeks of first pregnancy, whose severe HT and life-threatening symptoms prompted an emergency delivery without previous confirmation or medical treatment of a suspected PPGL. After confirmation, a right adrenal PPGL was surgically resected 4 months later, following 15 days of medical therapy. Case 2 is a 22-year-old female at 18 weeks of pregnancy whose symptomatic PPGL was resected in the second trimester. A next-generation sequencing panel, including 23 PPGL-related genes, found no germline pathogenic variants (GPVs) in case 1 and an exon 1-4 germinative heterozygous deletion of the MAX gene in case 2. Despite the different medical approaches, both cases had satisfactory outcomes. Although uncommon, PPGL should be considered in the differential diagnosis of HT in pregnancy since missing the diagnosis and failing to introduce appropriate and timely treatment may lead to dramatic consequences for the mother and fetus. PPGL diagnosed during reproductive age is likely to result from GPV, prompting genetic investigation and counseling.
Collapse
Affiliation(s)
- Rafael Buck Giorgi
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Division of Endocrinology, Faculty of Medical Sciences and Health, Pontific Catholic University of São Paulo, Sorocaba, Brazil
| | - Priscila Teixeira Aroucha
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Thalissa M. Favreto
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Micaela F. Montero
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Julia M. F. Velloni
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Ilana Korkes
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | | | - Caroline Olivati
- Research and Development Division, Fleury Group, São Paulo, Brazil
| | - Jose Viana Lima
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Research and Development Division, Fleury Group, São Paulo, Brazil
- Division of Endocrinology and Metabolism, Department of Medicine, Santa Casa de São Paulo, São Paulo, Brazil
| | - Claudio E. Kater
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Flavia A. Costa-Barbosa
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Research and Development Division, Fleury Group, São Paulo, Brazil
| |
Collapse
|
30
|
Ohmoto A, Hayashi N, Takahashi S, Ueki A. Current prospects of hereditary adrenal tumors: towards better clinical management. Hered Cancer Clin Pract 2024; 22:4. [PMID: 38532453 DOI: 10.1186/s13053-024-00276-6] [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: 08/27/2023] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Adrenocortical carcinoma (ACC) and pheochromocytoma/paraganglioma (PPGL) are two rare types of adrenal gland malignancies. Regarding hereditary tumors, some patients with ACC are associated with with Li-Fraumeni syndrome (LFS), and those with PPGL with multiple endocrine neoplasia type 2. Recent studies have expanded this spectrum to include other types of hereditary tumors, such as Lynch syndrome or familial adenomatous polyposis. Individuals harboring germline TP53 pathogenic variants that cause LFS have heterogeneous phenotypes depending on the respective variant type. As an example, R337H variant found in Brazilian is known as low penetrant. While 50-80% of pediatric ACC patients harbored a LFS, such a strong causal relationship is not observed in adult patients, which suggests different pathophysiologies between the two populations. As for PPGL, because multiple driver genes, such as succinate dehydrogenase (SDH)-related genes, RET, NF1, and VHL have been identified, universal multi-gene germline panel testing is warranted as a comprehensive and cost-effective approach. PPGL pathogenesis is divided into three molecular pathways (pseudohypoxia, Wnt signaling, and kinase signaling), and this classification is expected to result in personalized medicine based on genomic profiles. It remains unknown whether clinical characteristics differ between cases derived from genetic predisposition syndromes and sporadic cases, or whether the surveillance strategy should be changed depending on the genetic background or whether it should be uniform. Close cooperation among medical genomics experts, endocrinologists, oncologists, and early investigators is indispensable for improving the clinical management for multifaceted ACC and PPGL.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Division of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 417 East 68th Street, New York, NY, 10065, USA.
| | - Naomi Hayashi
- Division of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan
- Division of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan
| | - Shunji Takahashi
- Division of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan
- Division of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan
| | - Arisa Ueki
- Division of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 1358550, Japan
| |
Collapse
|
31
|
Mancini M, Buffet A, Porte B, Amar L, Lussey-Lepoutre C, Crinière L, Baudin E, Meatchi T, Gimenez-Roqueplo AP, Favier J, Burnichon N. EPAS1-mutated paragangliomas associated with haemoglobin disorders. Br J Haematol 2024; 204:1054-1060. [PMID: 38195958 DOI: 10.1111/bjh.19278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024]
Abstract
We report a large series of 40 patients presenting EPAS1-mutated paraganglioma (PGL) in whom we investigated a cause underlying chronic hypoxia. Four patients suffered from hypoxaemic heart disease. In patients with available haemoglobin electrophoresis results, 59% presented with a haemoglobin disorder, including six with sickle cell disease, five with sickle cell trait and two with heterozygous haemoglobin C disease. Histological and transcriptomic characterization of EPAS1 tumours revealed increased angiogenesis and high similarities with pseudohypoxic PGLs caused by VHL gene mutations. Sickle haemoglobinopathy carriers could thus be at increased risk for developing EPAS1-PGLs, which should be taken into account in their management and surveillance.
Collapse
Affiliation(s)
- Maxence Mancini
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Alexandre Buffet
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Baptiste Porte
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Laurence Amar
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Service d'Hypertension artérielle, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Charlotte Lussey-Lepoutre
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Service de Médecine Nucléaire, Sorbonne Université, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Lise Crinière
- Service d'endocrinologie, CHRU Bretonneau, Tours, France
| | - Eric Baudin
- Service de Médecine Nucléaire, Gustave Roussy, Villejuif, France
| | - Tchao Meatchi
- Service d'anatomie pathologique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Judith Favier
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Nelly Burnichon
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
32
|
Qin S, Xu Y, Yu S, Han W, Fan S, Ai W, Zhang K, Wang Y, Zhou X, Shen Q, Gong K, Sun L, Zhang Z. Molecular classification and tumor microenvironment characteristics in pheochromocytomas. eLife 2024; 12:RP87586. [PMID: 38407266 PMCID: PMC10942623 DOI: 10.7554/elife.87586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
Pheochromocytomas (PCCs) are rare neuroendocrine tumors that originate from chromaffin cells in the adrenal gland. However, the cellular molecular characteristics and immune microenvironment of PCCs are incompletely understood. Here, we performed single-cell RNA sequencing (scRNA-seq) on 16 tissues from 4 sporadic unclassified PCC patients and 1 hereditary PCC patient with Von Hippel-Lindau (VHL) syndrome. We found that intra-tumoral heterogeneity was less extensive than the inter-individual heterogeneity of PCCs. Further, the unclassified PCC patients were divided into two types, metabolism-type (marked by NDUFA4L2 and COX4I2) and kinase-type (marked by RET and PNMT), validated by immunohistochemical staining. Trajectory analysis of tumor evolution revealed that metabolism-type PCC cells display phenotype of consistently active metabolism and increased metastasis potential, while kinase-type PCC cells showed decreased epinephrine synthesis and neuron-like phenotypes. Cell-cell communication analysis showed activation of the annexin pathway and a strong inflammation reaction in metabolism-type PCCs and activation of FGF signaling in the kinase-type PCC. Although multispectral immunofluorescence staining showed a lack of CD8+ T cell infiltration in both metabolism-type and kinase-type PCCs, only the kinase-type PCC exhibited downregulation of HLA-I molecules that possibly regulated by RET, suggesting the potential of combined therapy with kinase inhibitors and immunotherapy for kinase-type PCCs; in contrast, the application of immunotherapy to metabolism-type PCCs (with antigen presentation ability) is likely unsuitable. Our study presents a single-cell transcriptomics-based molecular classification and microenvironment characterization of PCCs, providing clues for potential therapeutic strategies to treat PCCs.
Collapse
Affiliation(s)
- Sen Qin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Yawei Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Shimiao Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Wencong Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Shiheng Fan
- Shenzhen Institute of Ladder for Cancer ResearchShenzhenChina
| | - Wenxiang Ai
- Shenzhen Institute of Ladder for Cancer ResearchShenzhenChina
| | - Kenan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Yizhou Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Xuehong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Qi Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Kan Gong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Luyang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| | - Zheng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Department of Urology, Peking University First Hospital, Peking University Health Science CenterBeijingChina
| |
Collapse
|
33
|
Seabrook A, Vasudevan A, Neville K, Gerstl B, Benn D, Smith J, Kirk J, Gill A, Clifton-Bligh R, Tucker K. Genotype-phenotype correlations in paediatric and adolescent phaeochromocytoma and paraganglioma: a cross-sectional study. Arch Dis Child 2024; 109:201-208. [PMID: 38071512 DOI: 10.1136/archdischild-2023-325419] [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: 04/04/2023] [Accepted: 10/16/2023] [Indexed: 02/21/2024]
Abstract
Phaeochromocytoma (PC) and paraganglioma (PGL) syndromes associated with germline pathogenic variants are associated with high morbidity and mortality. Establishing genotype-phenotype correlations within a young population is challenging due to their rare occurrence. OBJECTIVE To describe genotype-phenotype correlations in paediatric and adolescent patients diagnosed with PC/PGL. To establish the incidence of PC/PGL in a young population and prevalence of germline pathogenic variants within this group. STUDY DESIGN We conducted a cross-sectional study of patients diagnosed with a PC/PGL aged 0-21 years old who were reviewed within Familial Cancer Services within New South Wales and the Australian Capital Territory, Australia. RESULTS A germline pathogenic variant was detected in 80% (24/30) of patients; SDHB: n=12, VHL: n=11, and MAX: n=1. Only patients harbouring a germline pathogenic variant reported a family history of syndromic tumours, those with apparently sporadic disease did not (62.5% versus 0%, p=0.02). All patients with VHL presented with an adrenal tumour compared with 25% of those with SDHB (100% versus 25%, p=0.01). Occurrence of multiple primary PC/PGL was seen in patients with VHL however was absent in patients with SDHB (36% versus 0%, p=0.03). Incidence rate of paediatric PC/PGL was 0.45 cases per million person years. CONCLUSIONS PC/PGL diagnosed in children and adolescents were strongly associated with germline pathogenic variants in VHL or SDHB. These patients should be referred to specialist services for family counselling and genetic testing along followed by investigations for the detection of bilateral, multifocal or metastatic disease, and lifelong surveillance for recurrent disease.
Collapse
Affiliation(s)
- Amanda Seabrook
- Cancer Genetics Diagnostic Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anand Vasudevan
- Deaprtment of Clinical Genetics, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Kristen Neville
- Department of Endocrinology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia
- Faculty of Medicine, School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Brigitte Gerstl
- The Kids Cancer Centre, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Diana Benn
- Cancer Genetics Diagnostic Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Janine Smith
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Clinical Genetics, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Judy Kirk
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Familial Cancer Service, Westmead Hospital, Westmead, New South Wales, Australia
| | - Anthony Gill
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Roderick Clifton-Bligh
- Cancer Genetics Diagnostic Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Katherine Tucker
- Hereditary Cancer Service, Prince of Wales Hospital Nelune Comprehensive Cancer Centre, Randwick, New South Wales, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
34
|
White G, Nonaka D, Chung TT, Oakey RJ, Izatt L. Somatic EPAS1 Variants in Pheochromocytoma and Paraganglioma in Patients With Sickle Cell Disease. J Clin Endocrinol Metab 2023; 108:3302-3310. [PMID: 37285480 PMCID: PMC10655516 DOI: 10.1210/clinem/dgad311] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/09/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
CONTEXT Somatic EPAS1 variants account for 5% to 8% of all pheochromocytoma and paragangliomas (PPGL) but are detected in over 90% of PPGL in patients with congenital cyanotic heart disease, where hypoxemia may select for EPAS1 gain-of-function variants. Sickle cell disease (SCD) is an inherited hemoglobinopathy associated with chronic hypoxia and there are isolated reports of PPGL in patients with SCD, but a genetic link between the conditions has yet to be established. OBJECTIVE To determine the phenotype and EPAS1 variant status of patients with PPGL and SCD. METHODS Records of 128 patients with PPGL under follow-up at our center from January 2017 to December 2022 were screened for SCD diagnosis. For identified patients, clinical data and biological specimens were obtained, including tumor, adjacent non-tumor tissue and peripheral blood. Sanger sequencing of exons 9 and 12 of EPAS1, followed by amplicon next-generation sequencing of identified variants was performed on all samples. RESULTS Four patients with both PPGL and SCD were identified. Median age at PPGL diagnosis was 28 years. Three tumors were abdominal paragangliomas and 1 was a pheochromocytoma. No germline pathogenic variants in PPGL-susceptibility genes were identified in the cohort. Genetic testing of tumor tissue detected unique EPAS1 variants in all 4 patients. Variants were not detected in the germline, and 1 variant was detected in lymph node tissue of a patient with metastatic disease. CONCLUSION We propose that somatic EPAS1 variants may be acquired through exposure to chronic hypoxia in SCD and drive PPGL development. Future work is needed to further characterize this association.
Collapse
Affiliation(s)
- Gemma White
- Department of Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Department of Clinical Genetics, Guy's and St Thomas’ NHS Foundation Trust, London, SE1 9RT, UK
| | - Daisuke Nonaka
- Department of Pathology, Guy's and St Thomas’ NHS Foundation Trust, London, SE1 7EH, UK
- Department of Cellular Pathology, King's College London, London, SE1 1UL, UK
| | - Teng-Teng Chung
- Department of Endocrinology, University College London Hospital NHS Foundation Trust, London, NW1 2BU, UK
| | - Rebecca J Oakey
- Department of Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Louise Izatt
- Department of Medical and Molecular Genetics, King's College London, London, SE1 9RT, UK
- Department of Clinical Genetics, Guy's and St Thomas’ NHS Foundation Trust, London, SE1 9RT, UK
| |
Collapse
|
35
|
Kiriakopoulos A, Giannakis P, Menenakos E. Pheochromocytoma: a changing perspective and current concepts. Ther Adv Endocrinol Metab 2023; 14:20420188231207544. [PMID: 37916027 PMCID: PMC10617285 DOI: 10.1177/20420188231207544] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
This article aims to review current concepts in diagnosing and managing pheochromocytoma and paraganglioma (PPGL). Personalized genetic testing is vital, as 40-60% of tumors are linked to a known mutation. Tumor DNA should be sampled first. Next-generation sequencing is the best and most cost-effective choice and also helps with the expansion of current knowledge. Recent advancements have also led to the increased incorporation of regulatory RNA, metabolome markers, and the NETest in PPGL workup. PPGL presentation is highly volatile and nonspecific due to its multifactorial etiology. Symptoms mainly derive from catecholamine (CMN) excess or mass effect, primarily affecting the cardiovascular system. However, paroxysmal nature, hypertension, and the classic triad are no longer perceived as telltale signs. Identifying high-risk subjects and diagnosing patients at the correct time by using appropriate personalized methods are essential. Free plasma/urine catecholamine metabolites must be first-line examinations using liquid chromatography with tandem mass spectrometry as the gold standard analytical method. Reference intervals should be personalized according to demographics and comorbidity. The same applies to result interpretation. Threefold increase from the upper limit is highly suggestive of PPGL. Computed tomography (CT) is preferred for pheochromocytoma due to better cost-effectiveness and spatial resolution. Unenhanced attenuation of >10HU in non-contrast CT is indicative. The choice of extra-adrenal tumor imaging is based on location. Functional imaging with positron emission tomography/computed tomography and radionuclide administration improves diagnostic accuracy, especially in extra-adrenal/malignant or familial cases. Surgery is the mainstay treatment when feasible. Preoperative α-adrenergic blockade reduces surgical morbidity. Aggressive metastatic PPGL benefits from systemic chemotherapy, while milder cases can be managed with radionuclides. Short-term postoperative follow-up evaluates the adequacy of resection. Long-term follow-up assesses the risk of recurrence or metastasis. Asymptomatic carriers and their families can benefit from surveillance, with intervals depending on the specific gene mutation. Trials primarily focusing on targeted therapy and radionuclides are currently active. A multidisciplinary approach, correct timing, and personalization are key for successful PPGL management.
Collapse
Affiliation(s)
- Andreas Kiriakopoulos
- Department of Surgery, ‘Evgenidion Hospital’, National and Kapodistrian University of Athens School of Medicine, 5th Surgical Clinic, Papadiamantopoulou 20 Str, PO: 11528, Athens 11528, Greece
| | | | | |
Collapse
|
36
|
de la Fouchardière C, Haissaguerre M, Decaussin-Petrucci M, Renaudin K, Deschamps F, Mirallié E, Murez T, Pattou F, Rocher L, Savoie PH, Faron M, Taieb D, Tabarin A, Bertherat J, Gimenez-Roqueplo AP, Amar L, Baudin E, Libé R. [French recommendations for malignant pheochromocytomas and paragangliomas by the national ENDOCAN-COMETE network]. Bull Cancer 2023; 110:1063-1083. [PMID: 37573200 DOI: 10.1016/j.bulcan.2023.06.002] [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: 12/22/2022] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 08/14/2023]
Abstract
Pheochromocytomas and paragangliomas are rare neuroendocrine tumors, developed respectively in the adrenal medulla and in extra-adrenal locations. Their malignancy is defined by the presence of distant metastases. Forty percent of them are inherited and can be part of different hereditary syndromes. Their management is ensured in France by the multidisciplinary expert centers of the ENDOCAN-COMETE national network "Cancers of the Adrenal gland", certified by the National Cancer Institute and discussed within multidisciplinary team meetings. The diagnostic and therapeutic work-up must be standardized, based on an expert analysis of clinical symptoms, hormonal biological secretions, genetics, morphological and specific metabolic imaging. In the context of a heterogeneous survival sometimes beyond seven to ten years, therapeutic intervention must be justified. This is multidisciplinary and relies on surgery, interventional radiology, external or internal radiotherapy and medical treatments such as sunitinib or dacarbazine and temodal chemotherapy. The personalized approach based on functional imaging fixation status and genetics is progressing despite the extreme rarity of this disease.
Collapse
Affiliation(s)
| | - Magalie Haissaguerre
- CHU de Bordeaux, hôpital Haut Lévêque, service d'endocrinologie, centre coordonnateur ENDOCAN-COMETE, Pessac, France
| | | | - Karine Renaudin
- CHU de Nantes, hôpital Hôtel-Dieu, anatomo-pathologie, Nantes, France
| | - Fréderic Deschamps
- Gustave-Roussy Cancer Campus, département de radiologie interventionnelle, Villejuif, France
| | - Eric Mirallié
- CHU de Nantes, hôpital Hôtel-Dieu, chirurgie cancérologique, digestive et endocrinienne, Institut des maladies de l'appareil digestif, Nantes, France
| | - Thibaut Murez
- CHU de Montpellier, département d'urologie et transplantation rénale, Montpellier, France
| | - François Pattou
- CHRU de Lille, département de chirurgie endocrinienne et métabolique, Lille, France
| | - Laurence Rocher
- Université Paris-Saclay, BIOMAPS, hôpital Antoine-Béclère, service de radiologie, Clamart, France
| | - Pierre-Henri Savoie
- Hôpital d'instruction des Armées Sainte-Anne, service d'urologie, Toulon, France
| | - Matthieu Faron
- Gustave-Roussy Cancer Campus, service de chirurgie viscérale oncologique, Villejuif, France
| | - David Taieb
- La Timone University Hospital, CERIMED, Aix-Marseille University, département de médecine nucléaire, Marseille, France
| | - Antoine Tabarin
- CHU de Bordeaux, hôpital Haut Lévêque, service d'endocrinologie, centre coordonnateur ENDOCAN-COMETE, Pessac, France
| | - Jérôme Bertherat
- Hôpital Cochin, CHU de Paris-Centre, service d'endocrinologie, centre coordonnateur ENDOCAN-COMETE, Paris, France
| | | | - Laurence Amar
- Hôpital européen Georges-Pompidou, service d'hypertension artérielle, Paris, France
| | - Eric Baudin
- Gustave-Roussy Cancer Campus, service de cancérologie endocrine, centre coordonnateur ENDOCAN-COMETE, Villejuif, France
| | - Rossella Libé
- Hôpital Cochin, CHU de Paris-Centre, service d'endocrinologie, centre coordonnateur ENDOCAN-COMETE, Paris, France.
| |
Collapse
|
37
|
Guo Q, Cheng ZM, Gonzalez-Cantú H, Rotondi M, Huelgas-Morales G, Ethiraj P, Qiu Z, Lefkowitz J, Song W, Landry BN, Lopez H, Estrada-Zuniga CM, Goyal S, Khan MA, Walker TJ, Wang E, Li F, Ding Y, Mulligan LM, Aguiar RCT, Dahia PLM. TMEM127 suppresses tumor development by promoting RET ubiquitination, positioning, and degradation. Cell Rep 2023; 42:113070. [PMID: 37659079 PMCID: PMC10637630 DOI: 10.1016/j.celrep.2023.113070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/06/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
The TMEM127 gene encodes a transmembrane protein of poorly known function that is mutated in pheochromocytomas, neural crest-derived tumors of adrenomedullary cells. Here, we report that, at single-nucleus resolution, TMEM127-mutant tumors share precursor cells and transcription regulatory elements with pheochromocytomas carrying mutations of the tyrosine kinase receptor RET. Additionally, TMEM127-mutant pheochromocytomas, human cells, and mouse knockout models of TMEM127 accumulate RET and increase its signaling. TMEM127 contributes to RET cellular positioning, trafficking, and lysosome-mediated degradation. Mechanistically, TMEM127 binds to RET and recruits the NEDD4 E3 ubiquitin ligase for RET ubiquitination and degradation via TMEM127 C-terminal PxxY motifs. Lastly, increased cell proliferation and tumor burden after TMEM127 loss can be reversed by selective RET inhibitors in vitro and in vivo. Our results define TMEM127 as a component of the ubiquitin system and identify aberrant RET stabilization as a likely mechanism through which TMEM127 loss-of-function mutations cause pheochromocytoma.
Collapse
Affiliation(s)
- Qianjin Guo
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Zi-Ming Cheng
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Hector Gonzalez-Cantú
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Matthew Rotondi
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Gabriela Huelgas-Morales
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Purushoth Ethiraj
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Zhijun Qiu
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Jonathan Lefkowitz
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Wan Song
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Bethany N Landry
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Hector Lopez
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Cynthia M Estrada-Zuniga
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Shivi Goyal
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Mohammad Aasif Khan
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Timothy J Walker
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Exing Wang
- Department Cell Structure and Anatomy, UTHSCSA, San Antonio, TX, USA
| | - Faqian Li
- Department of Pathology, UTHSCSA, San Antonio, TX, USA
| | - Yanli Ding
- Department of Pathology, UTHSCSA, San Antonio, TX, USA
| | - Lois M Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Ricardo C T Aguiar
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA; Mays Cancer Center, UTHSCSA, San Antonio, TX, USA; South Texas Veterans Health Care System, Audie Murphy VA Hospital, San Antonio, TX 78229, USA
| | - Patricia L M Dahia
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA; Mays Cancer Center, UTHSCSA, San Antonio, TX, USA.
| |
Collapse
|
38
|
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: 1] [Impact Index Per Article: 0.5] [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
|
39
|
Fagundes GFC, Freitas-Castro F, Santana LS, Afonso ACF, Petenuci J, Funari MFA, Guimaraes AG, Ledesma FL, Pereira MAA, Victor CR, Ferrari MSM, Coelho FMA, Srougi V, Tanno FY, Chambo JL, Latronico AC, Mendonca BB, Fragoso MCBV, Hoff AO, Almeida MQ. Evidence for a Founder Effect of SDHB Exon 1 Deletion in Brazilian Patients With Paraganglioma. J Clin Endocrinol Metab 2023; 108:2105-2114. [PMID: 36652439 DOI: 10.1210/clinem/dgad028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
CONTEXT Limited information is available concerning the genetic spectrum of pheochromocytoma and paraganglioma (PPGL) patients in South America. Germline SDHB large deletions are very rare worldwide, but most of the individuals harboring the SDHB exon 1 deletion originated from the Iberian Peninsula. OBJECTIVE Our aim was to investigate the spectrum of SDHB genetic defects in a large cohort of Brazilian patients with PPGLs. METHODS Genetic investigation of 155 index PPGL patients was performed by Sanger DNA sequencing, multiplex ligation-dependent probe amplification, and/or target next-generation sequencing panel. Common ancestrality was investigated by microsatellite genotyping with haplotype reconstruction, and analysis of deletion breakpoint. RESULTS Among 155 index patients, heterozygous germline SDHB pathogenic or likely pathogenic variants were identified in 22 cases (14.2%). The heterozygous SDHB exon 1 complete deletion was the most frequent genetic defect in SDHB, identified in 8 out of 22 (36%) of patients. Haplotype analysis of 5 SDHB flanking microsatellite markers demonstrated a significant difference in haplotype frequencies in a case-control permutation test (P = 0.03). More precisely, 3 closer/informative microsatellites were shared by 6 out of 8 apparently unrelated cases (75%) (SDHB-GATA29A05-D1S2826-D1S2644 | SDHB-186-130-213), which was observed in only 1 chromosome (1/42) without SDHB exon 1 deletion (X2 = 29.43; P < 0.001). Moreover, all cases with SDHB exon 1 deletion had the same gene breakpoint pattern of a 15 678 bp deletion previously described in the Iberian Peninsula, indicating a common origin. CONCLUSION The germline heterozygous SDHB exon 1 deletion was the most frequent genetic defect in the Brazilian PPGL cohort. Our findings demonstrated a founder effect for the SDHB exon 1 deletion in Brazilian patients with paragangliomas.
Collapse
Affiliation(s)
- Gustavo F C Fagundes
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Felipe Freitas-Castro
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Lucas S Santana
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Ana Caroline F Afonso
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Janaina Petenuci
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Mariana F A Funari
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Augusto G Guimaraes
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Felipe L Ledesma
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Maria Adelaide A Pereira
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Carolina R Victor
- Divisão de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brasil
- Centro de Oncologia Clínica, Rede D'Or, São Paulo 04543-000, Brasil
| | - Marcela S M Ferrari
- Divisão de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brasil
- Centro de Oncologia Clínica, Rede D'Or, São Paulo 04543-000, Brasil
| | - Fernando M A Coelho
- Instituto de Radiologia InRad, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Victor Srougi
- Divisão de Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Fabio Y Tanno
- Divisão de Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Jose L Chambo
- Divisão de Urologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Ana Claudia Latronico
- Unidade de Adrenal & Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Berenice B Mendonca
- Unidade de Adrenal & Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
| | - Maria Candida B V Fragoso
- Unidade de Adrenal & Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
- Divisão de Oncologia Endócrina, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brasil
| | - Ana O Hoff
- Centro de Oncologia Clínica, Rede D'Or, São Paulo 04543-000, Brasil
- Divisão de Oncologia Endócrina, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brasil
| | - Madson Q Almeida
- Unidade de Adrenal, Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brasil
- Divisão de Oncologia Endócrina, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brasil
| |
Collapse
|
40
|
Angeli SI, Chiossone K JA, Goncalves S, Telischi FF. Genotype-phenotype associations in paragangliomas of the temporal bone in a multi-ethnic cohort. Acta Otolaryngol 2023; 143:551-557. [PMID: 37354038 DOI: 10.1080/00016489.2023.2222149] [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: 03/20/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Temporal bone paragangliomas are rare tumours with variable presentation that can be hereditary. Identification of clinical and genetic factors of aggressive tumour behaviour is important. OBJECTIVE To determine the underlying genetic mutations and genotype/phenotype correlations in a multi-ethnic population of South Florida with sporadic temporal bone paragangliomas. METHODS In a cohort of glomus tympanicum (GT) and glomus jugulare (GJ) cases, we assessed the frequency of pathogenic single nucleotide variants, insertions, deletions, and duplications in coding exons of genes that have been associated with paragangliomas (SDHB, SDHC, SDHD, SDHA, SDHAF2, RET, NF1, VHL, TMEM127, and MAX). RESULTS None of the 12 GT cases had mutations. Among 13 GJ cases, we identified four mutation carriers (31%); two in SDHC, one in SDHB, and one in SDHD. All patients with pathogenic mutations were of Hispanic ethnicity, presented at a younger age (mean 27.5 versus 52.11 years), and with more advanced disease when compared to mutation-negative GJ cases.Conclusions and Significance: Mutations in the SDH genes are found in 31% of sporadic GJ. SDH-associated GJ had advanced disease and a 50% risk of metastasis. Our data supports emerging recommendations for genetic screening in all populations with GJ tumours as the genetic status informs management.
Collapse
Affiliation(s)
- Simon I Angeli
- University of Miami Miller School of Medicine, Otolaryngology, Miami, FL, USA
| | - Juan A Chiossone K
- University of Miami Miller School of Medicine, Otolaryngology, Miami, FL, USA
| | - Stefania Goncalves
- Neurotology Fellow, Jackson Memorial Hospital, Otolaryngology, Miami, FL, USA
| | - Fred F Telischi
- University of Miami Miller School of Medicine, Otolaryngology, Miami, FL, USA
| |
Collapse
|
41
|
Li M, He Y, Pang Y, Zhang J, Feng Y, He Y, Xu X, Wei Y, Zhong D, Deng W, Wang L, Yan B, Jiang Y, Xu N, Cai H, Wen Y, Ning J, Liu Y, Gao X, Shan Z, Liu L, Teng X, Richter S, Jiang J. Somatic IDH1 Hotspot Variants in Chinese Patients With Pheochromocytomas and Paragangliomas. J Clin Endocrinol Metab 2023; 108:1215-1223. [PMID: 36355572 DOI: 10.1210/clinem/dgac653] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022]
Abstract
CONTEXT IDH1 is a pheochromocytoma/paraganglioma (PPGL) susceptibility gene; however, its role, especially in the Chinese population, has not been characterized. OBJECTIVE To determine the prevalence of somatic IDH1 hotspot variants in a large cohort of Chinese patients with PPGLs and to summarize associated phenotypes. METHODS This retrospective cross-sectional study was based on a main cohort of 1141 patients with PPGLs from 2 tertiary-care centers in China. We included 50 cases with urinary bladder paragangliomas (UBPGLs), of whom 29 were part of the main cohort and 21 were from other centers. Two additional cases with IDH1 hotspot variants not part of the main cohort were also included for summarizing IDH1-associated phenotypes. Next-generation sequencing of tumor DNA was used to analyze a customized panel of genes. RESULTS The overall prevalence of IDH1 hotspot variants in the main cohort was 0.5% (6/1141). Among those PPGLs without mutations in 15 common driver genes, the prevalence of IDH1 variants was 0.9% (4/455). When restricted to paraganglioma (PGL) without mutations, the prevalence reached 4.7% (4/86). Among UBPGLs, IDH1 hotspot variants accounted for 8% (4/50). Together, all 10 patients (9 PGLs and 1 pheochromocytoma) with IDH1 hotspot variants, including 3 females with concurrent EPAS1 hotspot variants, had apparently sporadic tumors, without metastasis or recurrence. There were 3 patients with biochemical data, all showing a non-adrenergic phenotype. CONCLUSIONS The somatic IDH1 hotspot variants cause PPGL development in some Chinese patients, especially among those apparently sporadic PGLs with a non-adrenergic phenotype and without mutations in major PPGL driver genes.
Collapse
Affiliation(s)
- Minghao Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yazhi He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaowen Xu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yongbao Wei
- Shengli Clinical Medical College of Fujian Medical University and Department of Urology, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Dewen Zhong
- Department of Urology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Wanglong Deng
- Department of Urology, The First People's Hospital of Chenzhou, Chenzhou, 423000, China
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Bin Yan
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yazhuo Jiang
- Department of Urology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Ning Xu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 361003, China
| | - Hai Cai
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 361003, China
| | - Yanlin Wen
- Department of Urology, Nanchong Central Hospital, The Second Clinical Hospital of North Sichuan Medical College, Szechwan, 637000, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
42
|
Lui MS, Clemente-Gutierrez U, Skefos CM, Perrier ND. Succinate Dehydrogenase Mutations as Familial Pheochromocytoma Syndromes. Surg Oncol Clin N Am 2023; 32:289-301. [PMID: 36925186 DOI: 10.1016/j.soc.2022.10.006] [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: 03/17/2023]
Abstract
It is recognized that a large portion of pheochromocytoma and paraganglioma cases will have an underlying germline mutation, supporting the recommendation for universal genetic testing in all patients with PPGLs. A mutation in succinate dehydrogenase subunit B is associated with increased rates of developing synchronous and/or metachronous metastatic disease. Patients identified with this mutation require meticulous preoperative evaluation, a personalized surgical plan to minimize the risk of recurrence and tumor spread, and lifelong surveillance.
Collapse
Affiliation(s)
- Michael S Lui
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| | - Uriel Clemente-Gutierrez
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| | - Catherine M Skefos
- Clinical Cancer Genetics Program, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Nancy D Perrier
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW A vagal paraganglioma is a rare head and neck tumor arising from the paraganglionic tissue within the perineurium of the vagus nerve, anywhere along the course of the nerve. Due to its proximity to the internal carotid artery, the internal jugular vein and the lower cranial nerves, this disorder poses significant diagnostic and therapeutic challenges. The diagnostic workup and management keep on evolving. RECENT FINDINGS This article gives a concise update of the clinical spectrum and the current state-of-the-art diagnostic workup and management of vagal paraganglioma. SUMMARY Every patient with suspected vagal paraganglioma needs to be evaluated by a multidisciplinary team. The management strategy is selected depending on the growth rate of the tumor, the age and fitness of the patient, the number of affected cranial nerves, the metabolic activity of the paraganglioma, and the eventual multicentricity. An algorithm guiding the clinician through the different treatment options is presented.
Collapse
|
44
|
Yoshihama K, Mutai H, Sekimizu M, Ito F, Saito S, Nakamura S, Mikoshiba T, Nagai R, Takebayashi A, Miya F, Kosaki K, Ozawa H, Matsunaga T. Molecular basis of carotid body tumor and associated clinical features in Japan identified by genomic, immunohistochemical, and clinical analyses. Clin Genet 2023; 103:466-471. [PMID: 36597280 DOI: 10.1111/cge.14294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Carotid body tumor (CBT) is classified as a paraganglioma (PGL). Here, we report the genetic background, protein expression pattern, and clinical findings of 30 Japanese CBT cases. Germline pathogenic or likely pathogenic (P/LP) variants of genes encoding succinate dehydrogenase subunits (SDHs) were detected in 15 of 30 cases (50%). The SDHB variants were the most frequently detected, followed by SDHA and SDHD variants. One case with SDHAF2 variant was bilateral CBT, and other two multiple PGL cases were not detected P/LP variants. The three cases with germline variants that could be tested did not have somatic P/LP variants of the same genes. Immunohistochemical analysis showed negative SDHB signals in CBT tissues in five cases with germline P/LP variants of SDHB, SDHD, or SDHA. In addition, SDHB signals in CBT tissues were negative in four of nine cases without germline P/LP variants of SDHs. These findings suggest the involvement of unidentified molecular mechanisms affecting SDHs.
Collapse
Affiliation(s)
- Keisuke Yoshihama
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Hideki Mutai
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Mariko Sekimizu
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Fumihiro Ito
- Department of Otolaryngology, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Shin Saito
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Nakamura
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takuya Mikoshiba
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryoto Nagai
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akiko Takebayashi
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ozawa
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
- Department of Otolaryngology, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
- Medical Genetics Center, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| |
Collapse
|
45
|
Branzoli F, Salgues B, Marjańska M, Laloi-Michelin M, Herman P, Le Collen L, Delemer B, Riancho J, Kuhn E, Jublanc C, Burnichon N, Amar L, Favier J, Gimenez-Roqueplo AP, Buffet A, Lussey-Lepoutre C. SDHx mutation and pituitary adenoma: can in vivo 1H-MR spectroscopy unravel the link? Endocr Relat Cancer 2023; 30:ERC-22-0198. [PMID: 36449569 PMCID: PMC9885742 DOI: 10.1530/erc-22-0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
Germline mutations in genes encoding succinate dehydrogenase (SDH) are frequently involved in pheochromocytoma/paraganglioma (PPGL) development and were implicated in patients with the '3PAs' syndrome (associating pituitary adenoma (PA) and PPGL) or isolated PA. However, the causality link between SDHx mutation and PA remains difficult to establish, and in vivo tools for detecting hallmarks of SDH deficiency are scarce. Proton magnetic resonance spectroscopy (1H-MRS) can detect succinate in vivo as a biomarker of SDHx mutations in PGL. The objective of this study was to demonstrate the causality link between PA and SDH deficiency in vivo using 1H-MRS as a novel noninvasive tool for succinate detection in PA. Three SDHx-mutated patients suffering from a PPGL and a macroprolactinoma and one patient with an apparently sporadic non-functioning pituitary macroadenoma underwent MRI examination at 3 T. An optimized 1H-MRS semi-LASER sequence (TR = 2500 ms, TE = 144 ms) was employed for the detection of succinate in vivo. Succinate and choline-containing compounds were identified in the MR spectra as single resonances at 2.44 and 3.2 ppm, respectively. Choline compounds were detected in all the tumors (three PGL and four PAs), while a succinate peak was only observed in the three macroprolactinomas and the three PGL of SDHx-mutated patients, demonstrating SDH deficiency in these tumors. In conclusion, the detection of succinate by 1H-MRS as a hallmark of SDH deficiency in vivo is feasible in PA, laying the groundwork for a better understanding of the biological link between SDHx mutations and the development of these tumors.
Collapse
Affiliation(s)
- Francesca Branzoli
- Paris Brain Institute - Institut du Cerveau (ICM), Center for Neuroimaging Research (CENIR), Paris, France
- Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, Paris, France
| | - Betty Salgues
- Sorbonne University, nuclear medicine department, Pitié-Salpêtrière Hospital, Assistance -Publique Hôpitaux de Paris, Paris, France
- Paris Cardiovascular Research Center (PARCC), Inserm, Paris, France
| | - Małgorzata Marjańska
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Marie Laloi-Michelin
- Endocrinology department, Lariboisière Hospital, Assistance -Publique Hôpitaux de Paris, Paris, France
| | - Philippe Herman
- ENT unit, Lariboisière Hospital, Assistance -Publique Hôpitaux de Paris, Paris-Cité University, INSERM U1141, Paris, France
| | - Lauriane Le Collen
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, University of Lille, Lille, France
- Department of Endocrinology Diabetology, University Hospital Center of Reims, Reims, France
- Department of Genetic, University Hospital Center of Reims, Reims, France
| | - Brigitte Delemer
- Department of Endocrinology Diabetology, University Hospital Center of Reims, Reims, France
- CRESTIC EA 3804, University of Reims Champagne Ardenne, UFR Sciences Exactes et Naturelles, Moulin de La Housse, BP 1039, Reims, France
| | - Julien Riancho
- AP-HP, Hôpital Européen Georges Pompidou, Hypertension Unit, and Reference centre for rare adrenal diseases, Paris, France
| | - Emmanuelle Kuhn
- Pituitary Unit, Pitié-Salpêtrière Hospital APHP, Sorbonne University, Paris, France
| | - Christel Jublanc
- Pituitary Unit, Pitié-Salpêtrière Hospital APHP, Sorbonne University, Paris, France
| | - Nelly Burnichon
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Cité, Inserm, PARCC, Paris, France
| | - Laurence Amar
- AP-HP, Hôpital Européen Georges Pompidou, Hypertension Unit, and Reference centre for rare adrenal diseases, Paris, France
- Université Paris Cité, Inserm, PARCC, Paris, France
| | | | - Anne-Paule Gimenez-Roqueplo
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Cité, Inserm, PARCC, Paris, France
| | - Alexandre Buffet
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Cité, Inserm, PARCC, Paris, France
| | - Charlotte Lussey-Lepoutre
- Sorbonne University, nuclear medicine department, Pitié-Salpêtrière Hospital, Assistance -Publique Hôpitaux de Paris, Paris, France
- Paris Cardiovascular Research Center (PARCC), Inserm, Paris, France
| |
Collapse
|
46
|
Mellid S, Gil E, Letón R, Caleiras E, Honrado E, Richter S, Palacios N, Lahera M, Galofré JC, López-Fernández A, Calatayud M, Herrera-Martínez AD, Galvez MA, Matias-Guiu X, Balbín M, Korpershoek E, Lim ES, Maletta F, Lider S, Fliedner SMJ, Bechmann N, Eisenhofer G, Canu L, Rapizzi E, Bancos I, Robledo M, Cascón A. Co-occurrence of mutations in NF1 and other susceptibility genes in pheochromocytoma and paraganglioma. Front Endocrinol (Lausanne) 2023; 13:1070074. [PMID: 36760809 PMCID: PMC9905101 DOI: 10.3389/fendo.2022.1070074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction The percentage of patients diagnosed with pheochromocytoma and paraganglioma (altogether PPGL) carrying known germline mutations in one of the over fifteen susceptibility genes identified to date has dramatically increased during the last two decades, accounting for up to 35-40% of PPGL patients. Moreover, the application of NGS to the diagnosis of PPGL detects unexpected co-occurrences of pathogenic allelic variants in different susceptibility genes. Methods Herein we uncover several cases with dual mutations in NF1 and other PPGL genes by targeted sequencing. We studied the molecular characteristics of the tumours with co-occurrent mutations, using omic tools to gain insight into the role of these events in tumour development. Results Amongst 23 patients carrying germline NF1 mutations, targeted sequencing revealed additional pathogenic germline variants in DLST (n=1) and MDH2 (n=2), and two somatic mutations in H3-3A and PRKAR1A. Three additional patients, with somatic mutations in NF1 were found carrying germline pathogenic mutations in SDHB or DLST, and a somatic truncating mutation in ATRX. Two of the cases with dual germline mutations showed multiple pheochromocytomas or extra-adrenal paragangliomas - an extremely rare clinical finding in NF1 patients. Transcriptional and methylation profiling and metabolite assessment showed an "intermediate signature" to suggest that both variants had a pathological role in tumour development. Discussion In conclusion, mutations affecting genes involved in different pathways (pseudohypoxic and receptor tyrosine kinase signalling) co-occurring in the same patient could provide a selective advantage for the development of PPGL, and explain the variable expressivity and incomplete penetrance observed in some patients.
Collapse
Affiliation(s)
- Sara Mellid
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Gil
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rocío Letón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Susan Richter
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nuria Palacios
- Endocrinology Department, University Hospital Puerta de Hierro, Madrid, Spain
| | - Marcos Lahera
- Endocrinology and Nutrition Department, La Princesa University Hospital, Madrid, Spain
| | - Juan C. Galofré
- Department of Endocrinology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Adriá López-Fernández
- Hereditary Cancer Genetics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Calatayud
- Department of Endocrinology and Nutrition, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - María A. Galvez
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Bellvitge University Hospital, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Milagros Balbín
- Molecular Oncology Laboratory, Instituto Universitario de Oncologia del Principado de Asturias, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Esther Korpershoek
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Eugénie S. Lim
- Department of Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Francesca Maletta
- Pathology Unit , Department of Laboratory Medicine, Azienda Ospedaliero-Universitaria (AOU) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Sofia Lider
- Endocrinology Department, National Institute of Endocrinology, Bucharest, Romania
| | | | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Letizia Canu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Irina Bancos
- Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, United States
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
47
|
|
48
|
Tabebi M, Söderkvist P, Gimm O. Nuclear and mitochondrial DNA alterations in pheochromocytomas and paragangliomas, and their potential treatment. Endocr Relat Cancer 2023; 30:ERC-22-0217. [PMID: 36219865 DOI: 10.1530/erc-22-0217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Mitochondrial DNA (mtDNA) alterations have been reported in different types of cancers and are suggested to play important roles in cancer development and metastasis. However, there is little information about its involvement in pheochromocytomas and paragangliomas (PCCs/PGLs) formation. PCCs and PGLs are rare endocrine tumors of the chromaffin cells in the adrenal medulla and extra-adrenal paraganglia that can synthesize and secrete catecholamines. Over the last 3 decades, the genetic background of about 60% of PCCs/PGLs involving nuclear DNA alterations has been determined. Recently, a study showed that mitochondrial alterations can be found in around 17% of the remaining PCCs/PGLs. In this review, we summarize recent knowledge regarding both nuclear and mitochondrial alterations and their involvement in PCCs/PGLs. We also provide brief insights into the genetics and the molecular pathways associated with PCCs/PGLs and potential therapeutical targets.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Peter Söderkvist
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, Linköping University, Linköping, Sweden
| | - Oliver Gimm
- Department of Surgery, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| |
Collapse
|
49
|
|
50
|
Pacak K. New Biology of Pheochromocytoma and Paraganglioma. Endocr Pract 2022; 28:1253-1269. [PMID: 36150627 PMCID: PMC9982632 DOI: 10.1016/j.eprac.2022.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022]
Abstract
Pheochromocytomas and paragangliomas continue to be defined by significant morbidity and mortality despite their several recent advances in diagnosis, localization, and management. These adverse outcomes are largely related to mass effect as well as catecholamine-induced hypertension, tachyarrhythmias and consequent target organ damage, acute coronary syndromes, and strokes (ischemic and hemorrhagic stroke). Thus, a proper understanding of the physiology and pathophysiology of these tumors and recent advances are essential to affording optimal care. These major developments largely include a redefinition of metastatic behavior, a novel clinical categorization of these tumors into 3 genetic clusters, and an enhanced understanding of catecholamine metabolism and consequent specific biochemical phenotypes. Current advances in imaging of these tumors are shifting the paradigm from poorly specific anatomical modalities to more precise characterization of these tumors using the advent and development of functional imaging modalities. Furthermore, recent advances have revealed new molecular events in these tumors that are linked to their genetic landscape and, therefore, provide new therapeutic platforms. A few of these prospective therapies translated into new clinical trials, especially for patients with metastatic or inoperable tumors. Finally, outcomes are ever-improving as patients are cared for at centers with cumulative experience and well-established multidisciplinary tumor boards. In parallel, these centers have supported national and international collaborative efforts and worldwide clinical trials. These concerted efforts have led to improved guidelines collaboratively developed by healthcare professionals with a growing expertise in these tumors and consequently improving detection, prevention, and identification of genetic susceptibility genes in these patients.
Collapse
Affiliation(s)
- Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|