1
|
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
|
2
|
Prado Wohlwend S, Bello Arques P. Radio theranostics in paragangliomas and pheochromocytomas. Rev Esp Med Nucl Imagen Mol 2024; 43:500017. [PMID: 38735639 DOI: 10.1016/j.remnie.2024.500017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
This continuing education aims to present in a clear and easy-to-understand manner the biology of paragangliomas and pheochromocytomas (PPGLs), the functional imaging studies available for their diagnosis and therapeutic planning, the requirements necessary to administer radioligand therapy (RLT) and the characteristics of these treatments (inclusion criteria, administration protocols, adverse effects and future perspectives). In this pathology we have two RLT options: [131I]MIBG and [177Lu]Lu-DOTA-TATE. The indication for treatment is determined by the expression of its therapeutic target in functional imaging studies, allowing precision and personalized medicine. Although most of the results we have for both treatments have as origin small retrospective series, RLT is presented as a safe and well-tolerated therapeutic option in PPGLs with slow-moderate progression or with uncontrollable symptoms, obtaining high disease control rates.
Collapse
Affiliation(s)
- Stefan Prado Wohlwend
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Clinical Center of Excellence Pheo Para Alliance.
| | - Pilar Bello Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Clinical Center of Excellence Pheo Para Alliance
| |
Collapse
|
3
|
Taïeb D, Nölting S, Perrier ND, Fassnacht M, Carrasquillo JA, Grossman AB, Clifton-Bligh R, Wanna GB, Schwam ZG, Amar L, Bourdeau I, Casey RT, Crona J, Deal CL, Del Rivero J, Duh QY, Eisenhofer G, Fojo T, Ghayee HK, Gimenez-Roqueplo AP, Gill AJ, Hicks R, Imperiale A, Jha A, Kerstens MN, de Krijger RR, Lacroix A, Lazurova I, Lin FI, Lussey-Lepoutre C, Maher ER, Mete O, Naruse M, Nilubol N, Robledo M, Sebag F, Shah NS, Tanabe A, Thompson GB, Timmers HJLM, Widimsky J, Young WJ, Meuter L, Lenders JWM, Pacak K. Management of phaeochromocytoma and paraganglioma in patients with germline SDHB pathogenic variants: an international expert Consensus statement. Nat Rev Endocrinol 2024; 20:168-184. [PMID: 38097671 DOI: 10.1038/s41574-023-00926-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 02/17/2024]
Abstract
Adult and paediatric patients with pathogenic variants in the gene encoding succinate dehydrogenase (SDH) subunit B (SDHB) often have locally aggressive, recurrent or metastatic phaeochromocytomas and paragangliomas (PPGLs). Furthermore, SDHB PPGLs have the highest rates of disease-specific morbidity and mortality compared with other hereditary PPGLs. PPGLs with SDHB pathogenic variants are often less differentiated and do not produce substantial amounts of catecholamines (in some patients, they produce only dopamine) compared with other hereditary subtypes, which enables these tumours to grow subclinically for a long time. In addition, SDHB pathogenic variants support tumour growth through high levels of the oncometabolite succinate and other mechanisms related to cancer initiation and progression. As a result, pseudohypoxia and upregulation of genes related to the hypoxia signalling pathway occur, promoting the growth, migration, invasiveness and metastasis of cancer cells. These factors, along with a high rate of metastasis, support early surgical intervention and total resection of PPGLs, regardless of the tumour size. The treatment of metastases is challenging and relies on either local or systemic therapies, or sometimes both. This Consensus statement should help guide clinicians in the diagnosis and management of patients with SDHB PPGLs.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, Aix-Marseille University, La Timone University Hospital, Marseille, France
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nancy D Perrier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Jorge A Carrasquillo
- Molecular Imaging and Therapy Service, Radiology Department, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- NET Unit, Royal Free Hospital, London, UK
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital and Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - George B Wanna
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zachary G Schwam
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laurence Amar
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Hypertension Unit, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Ruth T Casey
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Joakim Crona
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cheri L Deal
- Research Center, CHU Sainte-Justine and Dept. of Paediatrics, University of Montreal, Montreal, Québec, Canada
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Quan-Yang Duh
- Department of Surgery, UCSF-Mount Zion, San Francisco, CA, USA
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Tito Fojo
- Columbia University Irving Medical Center, New York City, NY, USA
- James J. Peters VA Medical Center, New York City, NY, USA
| | - Hans K Ghayee
- Division of Endocrinology & Metabolism, Department of Medicine, University of Florida, Gainesville, FL, USA
- Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Department of Oncogenetics and Cancer Genomic Medicine, AP-HP, Hôpital européen Georges Pompidou, Paris, France
| | - Antony J Gill
- University of Sydney, Sydney NSW Australia, Cancer Diagnosis and Pathology Group Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- NSW Health Pathology Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Rodney Hicks
- Department of Medicine, St Vincent's Hospital Medical School, Melbourne, Victoria, Australia
| | - Alessio Imperiale
- Department of Nuclear Medicine and Molecular Imaging - Institut de Cancérologie de Strasbourg Europe (ICANS), IPHC, UMR 7178, CNRS, University of Strasbourg, Strasbourg, France
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michiel N Kerstens
- Department of Endocrinology, University Medical Center Groningen, Groningen, Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Máxima Center for paediatric oncology, Utrecht, Netherlands
| | - André Lacroix
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Canada
| | - Ivica Lazurova
- Department of Internal Medicine 1, University Hospital, P.J. Šafárik University, Košice, Slovakia
| | - Frank I Lin
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charlotte Lussey-Lepoutre
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
- Sorbonne University, Department of Nuclear Medicine, Pitié-Salpêtrière, Paris, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ozgur Mete
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mitsuhide Naruse
- Clinical Research Institute of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center and Endocrine Center, Kyoto, Japan
- Clinical Research Center, Ijinkai Takeda General Hospital, Kyoto, Japan
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Frédéric Sebag
- Department of Endocrine Surgery, Aix-Marseille University, Conception Hospital, Marseille, France
| | - Nalini S Shah
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Akiyo Tanabe
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine, Tokyo, Japan
| | - Geoffrey B Thompson
- Division of Endocrine Surgery, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jiri Widimsky
- Third Department of Medicine, Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - William J Young
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Leah Meuter
- Stanford University School of Medicine, Department of Physician Assistant Studies, Stanford, CA, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
4
|
Giraudet AL. [Combination of internal and external beam radiotherapy]. Cancer Radiother 2023; 27:754-758. [PMID: 37953187 DOI: 10.1016/j.canrad.2023.08.005] [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: 05/30/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 11/14/2023]
Abstract
External beam radiation therapy and internal vectorized radiation therapy are two types of radiotherapy that can be used to treat cancer. They differ in the way they are administered, and the type of radiation used. Although they can be effective in treating cancer, they each have their own advantages and disadvantages, and their combination could be synergistic. Preclinical studies on combined internal and external beam radiation therapy have mainly used radiolabelled antibodies, whose bone marrow toxicity remains the limiting factor in increasing the administered activities. The use of small radioligands in clinical trials has shown to be better tolerated and more effective, which explains their rapid development. The results of preclinical studies on combined internal and external beam radiation therapy appear heterogeneous, making it impossible to determine an ideal therapeutic sequencing scheme, and complicating the transposition to clinical studies. The few clinical studies on combined internal and external beam radiation therapy available to date have demonstrated feasibility and tolerability. More work remains to be done in the fields of dosimetry and radiobiology, as well as in the sequencing of these two irradiation modalities to optimize their combination.
Collapse
Affiliation(s)
- A-L Giraudet
- Centre Léon-Bérard, 15, rue Gabriel-Sarrazin, 69008 Lyon, France.
| |
Collapse
|
5
|
Fischer A, Kloos S, Remde H, Dischinger U, Pamporaki C, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Bechmann N, Hantel C, Mohr H, Pellegata NS, Bornstein SR, Kroiss M, Auernhammer CJ, Reincke M, Pacak K, Grossman AB, Beuschlein F, Nölting S. Responses to systemic therapy in metastatic pheochromocytoma/paraganglioma: a retrospective multicenter cohort study. Eur J Endocrinol 2023; 189:546-565. [PMID: 37949483 DOI: 10.1093/ejendo/lvad146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/02/2023] [Accepted: 09/19/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The therapeutic options for metastatic pheochromocytomas/paragangliomas (mPPGLs) include chemotherapy with cyclophosphamide/vincristine/dacarbazine (CVD), temozolomide monotherapy, radionuclide therapies, and tyrosine kinase inhibitors such as sunitinib. The objective of this multicenter retrospective study was to evaluate and compare the responses of mPPGLs including those with pathogenic variants in succinate dehydrogenase subunit B (SDHB), to different systemic treatments. DESIGN This is a retrospective analysis of treatment responses of mPPGL patients (n = 74) to systemic therapies. METHODS Patients with mPPGLs treated at 6 specialized national centers were selected based on participation in the ENSAT registry. Survival until detected progression (SDP) and disease-control rates (DCRs) at 3 months were evaluated based on imaging reports. RESULTS For the group of patients with progressive disease at baseline (83.8% of 74 patients), the DCR with first-line CVD chemotherapy was 75.0% (n = 4, SDP 11 months; SDHB [n = 1]: DCR 100%, SDP 30 months), with somatostatin peptide receptor-based radionuclide therapy (PPRT) 85.7% (n = 21, SDP 17 months; SDHB [n = 10]: DCR 100%, SDP 14 months), with 131I-meta-iodobenzylguanidine (131I-MIBG) 82.6% (n = 23, SDP 43 months; SDHB [n = 4]: DCR 100%, SDP 24 months), with sunitinib 100% (n = 7, SDP 18 months; SDHB [n = 3]: DCR 100%, SDP 18 months), and with somatostatin analogs 100% (n = 4, SDP not reached). The DCR with temozolomide as second-line therapy was 60.0% (n = 5, SDP 10 months; SDHB [n = 4]: DCR 75%, SDP 10 months). CONCLUSIONS We demonstrate in a real-life clinical setting that all current therapies show reasonable efficacy in preventing disease progression, and this is equally true for patients with germline SDHB mutations.
Collapse
Affiliation(s)
- Alessa Fischer
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Simon Kloos
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Hanna Remde
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ulrich Dischinger
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Institute de Salud Carlos III, Madrid, Spain
| | - Stephanie M J Fliedner
- First Department of Medicine, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Katharina Wang
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Julian Maurer
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Astrid Reul
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse, Dresden, Germany
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Stefan R Bornstein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christoph J Auernhammer
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Reincke
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD, United States
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, United Kingdom
- NET Unit, ENETS Center of Excellence, Royal Free Hospital, London, United Kingdom
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
6
|
Zhang X, Wakabayashi H, Hiromasa T, Kayano D, Kinuya S. Recent Advances in Radiopharmaceutical Theranostics of Pheochromocytoma and Paraganglioma. Semin Nucl Med 2023; 53:503-516. [PMID: 36641337 DOI: 10.1053/j.semnuclmed.2022.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
As a rare kind of non-epithelial neuroendocrine neoplasms, paragangliomas (PGLs) exhibit various clinical characteristics with excessive catecholamine secretion and have been a research focus in recent years. Although several modalities are available nowadays, radiopharmaceuticals play an integral role in the management of PGLs. Theranostics utilises radiopharmaceuticals for diagnostic and therapeutic intentions by aiming at a specific target in tumour and has been considered a possible means in diagnosis, staging, monitoring and treatment planning. Numerous radiopharmaceuticals have been developed over the past decades. 123/131-Metaiodobenzylguanidine (123/131I-MIBG), the theranostics pair target on norepinephrine transporter system, has remained a fantastic protocol for patients with PGLs because of disease control with limited toxicity. The high-specific-activity 131I-MIBG was authorised by the Food and Drug Administration as a systemic treatment method for metastatic PGLs in 2018. Afterward, peptide receptor radionuclide therapy, which uses radiolabelled somatostatin (SST) analogues, has been exploited as a superior substitute. 68Ga-somatostatin analogue (SSA) PET showed significant performance in diagnosing PGLs than MIBG scintigraphy, especially in patients with head and neck PGLs or SDHx mutation. 90Y/177Lu-DOTA-SSA is highly successful and has preserved favourable safety with mounting evidence regarding objective response, disease stabilisation, symptomatic and hormonal management and quality of life preservation. Besides the ordinary beta emitters, alpha-emitters such as 211At-MABG and 225Ac-DOTATATE have been investigated intensively in recent years. However, many studies are still in the pre-clinical stage, and more research is necessary. This review summarises the developments and recent advances in radiopharmaceutical theranostics of PGLs.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| |
Collapse
|
7
|
Carafone L, Victor A, Harbuz-Miller I. A SDHB Variant of Unknown Significance in a Patient With a Cardiac Functional Paraganglioma. JCEM CASE REPORTS 2023; 1:luad093. [PMID: 37908983 PMCID: PMC10580458 DOI: 10.1210/jcemcr/luad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Indexed: 11/02/2023]
Abstract
Cardiac paragangliomas are extremely rare tumors derived from chromaffin cells of the neural crest. Succinate dehydrogenase B (SDHB) mutations are associated with metastatic potential and potentially worse prognosis. Here we describe the case of a 64-year-old man who presented with chest pain, fatigue, and weight loss. Cardiac workup revealed a nearly 7-cm cardiac mass in the right lateral wall. Incisional biopsy demonstrated paraganglioma. Plasma free normetanephrine and chromogranin A were elevated. A DOTATATE positron emission tomography/computed tomography (PET/CT) revealed avidity of the mass with no evidence of distant metastases. Next-generation sequencing of the specimen demonstrated a variant of unknown significance of SDHB at H244D. Germline testing was negative. Surgical resection was aborted due to involvement of critical structures of the heart. Systemic treatment with the multi-tyrosine kinase inhibitor cabozantinib was initiated with subsequent improvements in biochemical markers as well as reductions in maximum standardized uptake value (SUVmax) on Ga-68 DOTATATE PET/CT. After 5 months of cabozantinib, he was unable to tolerate the side effects and external beam radiation therapy was completed. In this case, we report a novel somatic SDHB mutation at H244D in a sympathetic paraganglioma presenting as a cardiac mass.
Collapse
Affiliation(s)
- Lindsay Carafone
- Division of Endocrinology, University of Rochester, Rochester, NY 14642, USA
| | - Adrienne Victor
- Division of Hematology and Oncology, University of Rochester, Rochester, NY 14642, USA
| | - Inga Harbuz-Miller
- Division of Endocrinology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
8
|
Taïeb D, Wanna GB, Ahmad M, Lussey-Lepoutre C, Perrier ND, Nölting S, Amar L, Timmers HJLM, Schwam ZG, Estrera AL, Lim M, Pollom EL, Vitzthum L, Bourdeau I, Casey RT, Castinetti F, Clifton-Bligh R, Corssmit EPM, de Krijger RR, Del Rivero J, Eisenhofer G, Ghayee HK, Gimenez-Roqueplo AP, Grossman A, Imperiale A, Jansen JC, Jha A, Kerstens MN, Kunst HPM, Liu JK, Maher ER, Marchioni D, Mercado-Asis LB, Mete O, Naruse M, Nilubol N, Pandit-Taskar N, Sebag F, Tanabe A, Widimsky J, Meuter L, Lenders JWM, Pacak K. Clinical consensus guideline on the management of phaeochromocytoma and paraganglioma in patients harbouring germline SDHD pathogenic variants. Lancet Diabetes Endocrinol 2023; 11:345-361. [PMID: 37011647 PMCID: PMC10182476 DOI: 10.1016/s2213-8587(23)00038-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 04/05/2023]
Abstract
Patients with germline SDHD pathogenic variants (encoding succinate dehydrogenase subunit D; ie, paraganglioma 1 syndrome) are predominantly affected by head and neck paragangliomas, which, in almost 20% of patients, might coexist with paragangliomas arising from other locations (eg, adrenal medulla, para-aortic, cardiac or thoracic, and pelvic). Given the higher risk of tumour multifocality and bilaterality for phaeochromocytomas and paragangliomas (PPGLs) because of SDHD pathogenic variants than for their sporadic and other genotypic counterparts, the management of patients with SDHD PPGLs is clinically complex in terms of imaging, treatment, and management options. Furthermore, locally aggressive disease can be discovered at a young age or late in the disease course, which presents challenges in balancing surgical intervention with various medical and radiotherapeutic approaches. The axiom-first, do no harm-should always be considered and an initial period of observation (ie, watchful waiting) is often appropriate to characterise tumour behaviour in patients with these pathogenic variants. These patients should be referred to specialised high-volume medical centres. This consensus guideline aims to help physicians with the clinical decision-making process when caring for patients with SDHD PPGLs.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, Aix-Marseille University, La Timone University Hospital, Marseille, France
| | - George B Wanna
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maleeha Ahmad
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Charlotte Lussey-Lepoutre
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France; Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Nancy D Perrier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Svenja Nölting
- Svenja Nölting, Department of Endocrinology, Diabetology, and Clinical Nutrition, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Laurence Amar
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France; Unité d'hypertension artérielle, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Zachary G Schwam
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony L Estrera
- Department of Cardiothoracic and Vascular Surgery, UTHealth Houston, McGovern Medical School, Memorial Hermann Hospital Heart and Vascular Institute, Houston, TX, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Erqi Liu Pollom
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Lucas Vitzthum
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine and Research Center, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Ruth T Casey
- Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Frédéric Castinetti
- Department of Endocrinology, Aix-Marseille University, Conception University Hospital, Marseille, France; INSERM U1251, Aix-Marseille University, Conception University Hospital, Marseille, France
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney, NSW, Australia; Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Eleonora P M Corssmit
- Department of Endocrinology, Center of Endocrine Tumors Leiden, Leiden University Medical Centre, Leiden, Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hans K Ghayee
- Division of Endocrinology and Metabolism, Department of Medicine, Malcom Randall VA Medical Center, University of Florida, Gainesville, FL, USA
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, Inserm, PARCC, Equipe Labellisée par la Ligue contre le Cancer, Paris, France; Département de Médecine Génomique des Tumeurs et des Cancers, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ashley Grossman
- Green Templeton College, University of Oxford, Oxford, UK; NET Unit, Royal Free Hospital, London, UK
| | - Alessio Imperiale
- Department of Nuclear Medicine and Molecular Imaging, Institut de Cancérologie de Strasbourg Europe, IPHC, UMR 7178, CNRS, University of Strasbourg, Strasbourg, France
| | - Jeroen C Jansen
- Department of Otorhinolaryngology, Leiden University Medical Centre, Leiden, Netherlands
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michiel N Kerstens
- Department of Endocrinology, University Medical Center Groningen, Groningen, Netherlands
| | - Henricus P M Kunst
- Department of Otolaryngology and Head & Neck Surgery, Dutch Academic Alliance Skull Base Pathology, Radboud University Medical Center, Nijmegen, Netherlands; Department of Otolaryngology and Head & Neck Surgery, Dutch Academic Alliance Skull Base Pathology, Maastricht University Medical Center, Maastricht, Netherlands
| | - James K Liu
- Department of Neurosurgical Surgery, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Eamonn R Maher
- Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Daniele Marchioni
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital of Verona, Verona, Italy
| | - Leilani B Mercado-Asis
- Section of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine & Surgery, University of Santo Tomas Hospital, University of Santo Tomas, Manila, Philippines
| | - Ozgur Mete
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Endocrine Pathology Society, Toronto, ON, Canada
| | - Mitsuhide Naruse
- Medical Center and Endocrine Center, Ijinkai Takeda General Hospital, Kyoto, Japan
| | - Naris Nilubol
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Neeta Pandit-Taskar
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frédéric Sebag
- Department of Endocrine Surgery, Aix-Marseille University, Conception University Hospital, Marseille, France
| | - Akiyo Tanabe
- Division of Diabetes, Endocrinology, and Metabolism, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jiri Widimsky
- Third Department of Medicine, Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Leah Meuter
- Department of Physician Assistant Studies, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jacques W M Lenders
- Department of Medicine ΙΙI, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Gu Z, Hu K, Liang Y, Zhang F, Tong A, Hou X. Favorable outcome in advanced pheochromocytoma and paraganglioma after hypofractionated intensity modulated radiotherapy. J Endocrinol Invest 2023; 46:477-485. [PMID: 36121637 DOI: 10.1007/s40618-022-01908-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 08/19/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE The purpose of this study was to review outcomes of patients with advanced/metastatic pheochromocytoma/paraganglioma (PPGL) treated at our institution with Intensity-modulated radiotherapy (IMRT), describe the treatment outcomes, and determine predictors. METHODS A retrospective study on patients with advanced/metastatic PPGL who received IMRT at Peking Union Medical College Hospital between 2014 and 2019. A total of 14 patients with 17 lesions were included in this study. Ultra-hypofractionated radiation therapy was used for 7 lesions in 5 patients, while hypofractionated radiation therapy was used for 8 lesions in 7 patients. 2 patients got conventional fractionated radiotherapy. Patients who received external beam radiation therapy were given a median total radiation dose of 74.4/130 Gy (BED10/3) in a median of 13 fractions. RESULTS OS at 2 years was 78% for all patients. For lesions evaluated by RECIST response, at least stable disease of the target lesion was achieved in 94% and distant progression in 28.5%, with an average time to progression of 5.2 months. Patients with locally advanced primary tumors or recurred in situ (n = 8) achieved 100% local control, and none of them got recurrence or distant metastasis after radiotherapy at last follow-up (median 29 months). Of patients with catecholamine-related syndromes (n = 12), 91% of symptomatic lesions improved following radiation therapy and a more than 50% decline in catecholamines. CONCLUSIONS We have found hypofractionated IMRT effective as an additional therapy for patients with advanced primary tumors or recurrence in situ and not amenable to complete surgical resection.
Collapse
Affiliation(s)
- Z Gu
- Department of Radiation Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Dongdansantiao No. 5, Dongcheng District, Beijing, China
| | - K Hu
- Department of Radiation Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Dongdansantiao No. 5, Dongcheng District, Beijing, China
| | - Y Liang
- Department of Radiation Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Dongdansantiao No. 5, Dongcheng District, Beijing, China
| | - F Zhang
- Department of Radiation Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Dongdansantiao No. 5, Dongcheng District, Beijing, China
| | - A Tong
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - X Hou
- Department of Radiation Oncology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Dongdansantiao No. 5, Dongcheng District, Beijing, China.
| |
Collapse
|
10
|
Gan L, Shen XD, Ren Y, Cui HX, Zhuang ZX. Diagnostic features and therapeutic strategies for malignant paraganglioma in a patient: A case report. World J Clin Cases 2022; 10:9834-9844. [PMID: 36186170 PMCID: PMC9516940 DOI: 10.12998/wjcc.v10.i27.9834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/01/2022] [Accepted: 08/17/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Paragangliomas and extra-adrenal pheochromocytomas are uncommon neuroendocrine tumors with ubiquitous distribution. Malignant paraganglioma is a relatively rare entity. We report the treatment and pathological characteristics of a patient with malignant paraganglioma, and summarize the latest advances in the treatment of malignant paraganglioma based on a literature review.
CASE SUMMARY A 45-year-old Chinese woman presented to the hospital due to pain in the waist (right side) and right buttock, and was diagnosed as malignant paraganglioma after the placement of ureteral stent, implantation of ileus catheter, and transvaginal removal of the vaginal mass. After relief of intestinal obstruction, the patient received intravenous chemotherapy and peritoneal perfusion chemotherapy. Although her pelvic mass disease was stable, she developed multiple liver metastases and bone metastases. Due to the development of spinal cord compression, she underwent orthopedic surgery, followed by radiotherapy, and molecular targeted therapy with apatinib, but with poor disease control.
CONCLUSION Clinical management of paraganglioma is challenging for endocrinologists and oncologists. Prospective studies are required to develop standardized therapeutic strategies for malignant paragangliomas.
Collapse
Affiliation(s)
- Lei Gan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Xu-Dong Shen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Yang Ren
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Hong-Xia Cui
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Zhi-Xiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| |
Collapse
|
11
|
Zhang X, Wakabayashi H, Kayano D, Inaki A, Kinuya S. I-131 metaiodobenzylguanidine therapy is a significant treatment option for pheochromocytoma and paraganglioma. Nuklearmedizin 2022; 61:231-239. [PMID: 35668668 DOI: 10.1055/a-1759-2050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIM Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumours of chromaffin cells. Several modalities are currently available to treat patients with PPGL. These treatment modalities include surgery, chemotherapy, molecular targeted therapy and radiopharmaceuticals. METHODS I-131 metaiodobenzylguanidine (mIBG), a classic radiopharmaceutical, can be taken up through specific receptors and sited into many, but not all, PPGL cells. RESULTS Many studies have investigated the efficacy and toxicity of I-131 mIBG therapy. These studies reported significant results in terms of objective, hormonal and symptomatic responses as well as tolerable toxicities in patients. CONCLUSION This article reviews the reported experiences of patients who underwent I-131 mIBG therapy for PPGL with a focus on functions and deficiencies of the therapy.
Collapse
Affiliation(s)
- Xue Zhang
- Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | | | - Daiki Kayano
- Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Anri Inaki
- Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Seigo Kinuya
- Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
12
|
Lin EP, Chin BB, Fishbein L, Moritani T, Montoya SP, Ellika S, Newlands S. Head and Neck Paragangliomas: An Update on the Molecular Classification, State-of-the-Art Imaging, and Management Recommendations. Radiol Imaging Cancer 2022; 4:e210088. [PMID: 35549357 DOI: 10.1148/rycan.210088] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Paragangliomas are neuroendocrine tumors that derive from paraganglia of the autonomic nervous system, with the majority of parasympathetic paragangliomas arising in the head and neck. More than one-third of all paragangliomas are hereditary, reflecting the strong genetic predisposition of these tumors. The molecular basis of paragangliomas has been investigated extensively in the past couple of decades, leading to the discovery of several molecular clusters and more than 20 well-characterized driver genes (somatic and hereditary), which are more than are known for any other endocrine tumor. Head and neck paragangliomas are largely related to the pseudohypoxia cluster and have been previously excluded from most molecular profiling studies. This review article introduces the molecular classification of paragangliomas, with a focus on head and neck paragangliomas, and discusses its impact on the management of these tumors. Genetic testing is now recommended for all patients with paragangliomas to provide screening and surveillance recommendations for patients and relatives. While CT and MRI provide excellent anatomic characterization of paragangliomas, gallium 68 tetraazacyclododecane tetraacetic acid-octreotate (ie, 68Ga-DOTATATE) has superior sensitivity and is recommended as first-line imaging in patients with head and neck paragangliomas with concern for multifocal and metastatic disease, patients with known multifocal and metastatic disease, and in candidates for targeted peptide-receptor therapy. Keywords: Molecular Imaging, MR Perfusion, MR Spectroscopy, Neuro-Oncology, PET/CT, SPECT/CT, Head/Neck, Genetic Defects © RSNA, 2022.
Collapse
Affiliation(s)
- Edward P Lin
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Bennett B Chin
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Lauren Fishbein
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Toshio Moritani
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Simone P Montoya
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Shehanaz Ellika
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| | - Shawn Newlands
- From the Departments of Imaging Sciences (E.P.L., S.E.) and Otolaryngology (S.N.), University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642; Departments of Radiology (B.B.C.) and Medicine (L.F.), University of Colorado School of Medicine, Denver, Colo; Department of Radiology, University of Michigan, Ann Arbor, Mich (T.M.); Eastern Radiologists, East Carolina University, Vidant Medical Center, Greenville, NC (S.P.M.); and Department of Radiology, Massachusetts General Hospital, Boston, Mass (S.P.M.)
| |
Collapse
|
13
|
Inaki A, Shiga T, Tsushima Y, Jinguji M, Wakabayashi H, Kayano D, Akatani N, Yamase T, Kunita Y, Watanabe S, Hiromasa T, Mori H, Hirata K, Watanabe S, Higuchi T, Tomonaga H, Kinuya S. An open-label, single-arm, multi-center, phase II clinical trial of single-dose [ 131I]meta-iodobenzylguanidine therapy for patients with refractory pheochromocytoma and paraganglioma. Ann Nucl Med 2022; 36:267-278. [PMID: 34870794 PMCID: PMC8897386 DOI: 10.1007/s12149-021-01699-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/17/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE In this phase II study, we aimed to investigate the efficacy and safety of single-dose [131I]meta-iodobenzylguanidine (131I-mIBG) therapy in patients with refractory pheochromocytoma and paraganglioma (PPGL). PATIENTS AND METHODS This study was designed as an open-label, single-arm, multi-center, phase II clinical trial. The enrolled patients were administered 7.4 GBq of 131I-mIBG. Its efficacy was evaluated 12 and 24 weeks later, and its safety was monitored continuously until the end of the study. We evaluated the biochemical response rate as the primary endpoint using the one-sided exact binomial test based on the null hypothesis (≤ 5%). RESULTS Seventeen patients were enrolled in this study, of which 16 were treated. The biochemical response rate (≥ 50% decrease in urinary catecholamines) was 23.5% (90% confidence interval: 8.5-46.1%, p = 0.009). The radiographic response rates, determined with CT/MRI according to the response evaluation criteria in solid tumors (RECIST) version 1.1 and 123I-mIBG scintigraphy were 5.9% (0.3%-25.0%) and 29.4% (12.4%-52.2%), respectively. The most frequent non-hematologic treatment-emergent adverse events (TEAEs) were gastrointestinal symptoms including nausea, appetite loss, and constipation, which were, together, observed in 15 of 16 patients. Hematologic TEAEs up to grade 3 were observed in 14 of 16 patients. No grade 4 or higher TEAEs were observed. All patients had experienced at least one TEAE, but no fatal or irreversible TEAEs were observed. CONCLUSION A single dose 131I-mIBG therapy was well tolerated by patients with PPGL, and statistically significantly reduced catecholamine levels compared to the threshold response rate, which may lead to an improved prognosis for these patients.
Collapse
Affiliation(s)
- Anri Inaki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Tohru Shiga
- Department of Clinical Research and Trial, Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1-banchi Hikarigaoka, Fukushima, Fukushima 960-1295 Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma 371-8511 Japan
| | - Megumi Jinguji
- Department of Radiology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544 Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Norihito Akatani
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Takafumi Yamase
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Yuji Kunita
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Satoru Watanabe
- Department of Functional Imaging and Artificial Intelligence, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Hiroshi Mori
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, 5-chome Kita-14-jou, Kita-ku, Sapporo, Hokkaido 060-8648 Japan
| | - Shiro Watanabe
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, 5-chome Kita-14-jou, Kita-ku, Sapporo, Hokkaido 060-8648 Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma 371-8511 Japan
| | - Hiroyasu Tomonaga
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma 371-8511 Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| |
Collapse
|
14
|
Prado-Wohlwend S, del Olmo-García MI, Bello-Arques P, Merino-Torres JF. [ 177Lu]Lu-DOTA-TATE and [ 131I]MIBG Phenotypic Imaging-Based Therapy in Metastatic/Inoperable Pheochromocytomas and Paragangliomas: Comparative Results in a Single Center. Front Endocrinol (Lausanne) 2022; 13:778322. [PMID: 35197929 PMCID: PMC8859101 DOI: 10.3389/fendo.2022.778322] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/10/2022] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The aim of the study is to assess phenotypic imaging patterns and the response to treatment with [177Lu]Lu-DOTA-TATE and/or [131I]MIBG in paragangliomas (PGLs) and pheochromocytomas (PHEOs), globally and according to the primary location. METHODS This is a 17-patient retrospective observational study, with 9 cases treated with [177Lu]Lu-DOTA-TATE and 8 with [131I]MIBG (37 total treatments). Functional imaging scans and treatment responses were studied in order to choose the best therapeutic option and to define the progression-free survival (PFS) and disease control rate (DCR) according to treatment modality and primary location. RESULTS All patients were studied with phenotypic nuclear medicine images. Twelve of 17 patients were tested with both [123I]MIBG and somatostatin receptor images, and 6/12 showed appropriate expression of both targets to treatment in the phenotypic images. The rest of the patients were tested with one of the image modalities or only showed suitable uptake of a single radiotracer and were treated with the corresponding therapeutic option. [177Lu]Lu-DOTA-TATE PFS was 29 months with a DCR of 88.8%. [131I]MIBG PFS was 18.5 months with a 62.5% DCR. According to the primary location, the best PFS was in PHEOs treated with [177Lu]Lu-DOTA-TATE. Although the series are small due to the low disease prevalence and do not allow to yield statistically significant differences, this first study comparing [177Lu]Lu-DOTA-TATE and [131I]MIBG displays a trend to an overall longer PFS with [177Lu]Lu-DOTA-TATE, especially in the adrenal primary location. When both radionuclide targets are expressed, the patients' comorbidity and treatment effectiveness should be valued together with the intensity uptake in the phenotypic image in order to choose the best therapeutic option. These preliminary retrospective results reinforce the need for a prospective, multicentric trial to be confirmed.
Collapse
Affiliation(s)
- Stefan Prado-Wohlwend
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- *Correspondence: Stefan Prado-Wohlwend,
| | | | - Pilar Bello-Arques
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- Medicine Department, Universitat de València, Valencia, Spain
| |
Collapse
|
15
|
Prado-Wohlwend S, del Olmo-García MI, Bello-Arques P, Merino-Torres JF. Response to targeted radionuclide therapy with [ 131I]MIBG AND [ 177Lu]Lu-DOTA-TATE according to adrenal vs. extra-adrenal primary location in metastatic paragangliomas and pheochromocytomas: A systematic review. Front Endocrinol (Lausanne) 2022; 13:957172. [PMID: 36339441 PMCID: PMC9630737 DOI: 10.3389/fendo.2022.957172] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Targeted radionuclide therapy (TRT) with [131I]MIBG and [177Lu]Lu-DOTA-TATE is an alternative treatment to the classic schemes in slow progressive metastatic/inoperable paraganglioma (PGL) and pheochromocytoma (PHEO). There is no consensus on which treatment to administer and/or the best sequence in patients who are candidates for both therapies. To clarify these questions, this systematic review assesses the prognostic value of [131I]MIBG and [177Lu]Lu-DOTA-TATE (PRRT-Lu) treatments in terms of progression-free survival (PFS) both globally and considering the primary location. METHODS This review was developed according to the PRISMA Statement with 27 final studies (608 patients). Patient characteristics, treatment procedure, and follow-up criteria were evaluated. In addition, a Bayesian linear regression model weighted according to its sample size and an alternative model, which also included an interaction between the treatment and the proportion of PHEOs, were carried out, adjusted by a Student's t distribution. RESULTS In linear regression models, [131I]MIBG overall PFS was, on average, 10 months lower when compared with PRRT-Lu. When considering the interaction between treatment responses and the proportion of PHEOs, PRRT-Lu showed remarkably better results in adrenal location. The PFS of PRRT-Lu was longer when the ratio of PHEOs increased, with a decrease in [131I]MIBG PFS by 1.9 months for each 10% increase in the proportion of PHEOs in the sample. CONCLUSION Methodology, procedure, and PFS from the different studies are quite heterogeneous. PRRT-Lu showed better results globally and specifically in PHEOs. This fact opens the window to prospective trials comparing or sequencing [131I]MIBG and PRRT-Lu.
Collapse
Affiliation(s)
- Stefan Prado-Wohlwend
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- *Correspondence: Stefan Prado-Wohlwend,
| | | | - Pilar Bello-Arques
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- Medicine Department, Universitat de València, Valencia, Spain
| |
Collapse
|
16
|
Al-Dasuqi K, Irshaid L, Mathur M. Radiologic-Pathologic Correlation of Primary Retroperitoneal Neoplasms. Radiographics 2021; 40:1631-1657. [PMID: 33001785 DOI: 10.1148/rg.2020200015] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
An earlier incorrect version of this article appeared in print. The online version is correct.
Collapse
Affiliation(s)
- Khalid Al-Dasuqi
- From the Department of Radiology and Biomedical Imaging (K.A.D., M.M.) and Department of Pathology (L.I.), Yale School of Medicine, 333 Cedar St, PO Box 208042, Room TE-2, New Haven, CT 06520
| | - Lina Irshaid
- From the Department of Radiology and Biomedical Imaging (K.A.D., M.M.) and Department of Pathology (L.I.), Yale School of Medicine, 333 Cedar St, PO Box 208042, Room TE-2, New Haven, CT 06520
| | - Mahan Mathur
- From the Department of Radiology and Biomedical Imaging (K.A.D., M.M.) and Department of Pathology (L.I.), Yale School of Medicine, 333 Cedar St, PO Box 208042, Room TE-2, New Haven, CT 06520
| |
Collapse
|
17
|
Tu X, Zhang N, Zhuang X, Chen S, Luo X. Incidental diagnosis of nonfunctional bladder paraganglioma: a case report and literature review. BMC Urol 2021; 21:98. [PMID: 34238271 PMCID: PMC8265019 DOI: 10.1186/s12894-021-00863-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Nonfunctional bladder paragangliomas is a rare urological disease. It may present clinical, radiology and pathological features similar to bladder cancer, Only scarce reports have been reported. Urologist must identify this generally benign neuroendocrine neoplasm to avoid misdiagnosis. CASE PRESENTATION A 62-year-old female presented the outpatient department of our hospital with the symptoms of stomachache, frequent micturition, and urination pain for 20 days. Diagnosed with high blood pressure 1 year ago, administered Amlodipine besylate tablets 5 mg po qd occasionally, did not check blood pressure; denied any tumor observation in the family history. Color ultrasound of the urinary system showed a 38 mm × 34 mm hypoechoic mass on the right side of the bladder, CDFI: in the masses, blood supply was sufficient. Cystoscope showed bladder occupying lesion. Biopsy diagnosis: papillary polypoid cystitis was suspected as a malignant change (Fig. 3a). Then, the patient was admitted to our urological department. Further, computer tomography urography considered bladder cancer. Cystoscopy and biopsy failed to define the nature of the lesions in our outpatient department, which prompted a transurethral resection of the bladder tumor. histopathological and immunohistochemical results were diagnosed as bladder paragangliomas. For the reason, the tumor was removed by partial resection of the bladder. The postoperative recovery and follow-up were uneventful. CONCLUSIONS Nonfunctional bladder paragangliomas are occasionally found on imaging studies with the symptoms of urinary tract infection or/and intermittent painless hematuria. It may present clinical, radiology and pathological features similar to bladder cancer, so knowledge of this generally benign neuroendocrine neoplasm is of great importance to avoid misdiagnosis. It should be accompanied by the clinical and pathological characteristics of the patient and image changes. Partial resection of the bladder can effectively treat this disease.
Collapse
Affiliation(s)
- Xi Tu
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Neng Zhang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xiyao Zhuang
- Department of Internal Medicine, Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shulian Chen
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xu Luo
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
18
|
Suman SK, Subramanian S, Mukherjee A. Combination radionuclide therapy: A new paradigm. Nucl Med Biol 2021; 98-99:40-58. [PMID: 34029984 DOI: 10.1016/j.nucmedbio.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022]
Abstract
Targeted molecular radionuclide therapy (MRT) has shown its potential for the treatment of cancers of multiple origins. A combination therapy strategy employing two or more distinct therapeutic approaches in cancer management is aimed at circumventing tumor resistance by simultaneously targeting compensatory signaling pathways or bypassing survival selection mutations acquired in response to individual monotherapies. Combination radionuclide therapy (CRT) is a newer application of the concept, utilizing a combination of radiolabeled molecular targeting agents with chemotherapy and beam radiation therapy for enhanced therapeutic index. Encouraging results are reported with chemotherapeutic agents in combination with radiolabeled targeting molecules for cancer therapy. With increasing awareness of the various survival and stress response pathways activated after radionuclide therapy, different holistic combinations of MRT agents with radiosensitizers targeting such pathways have also been explored. MRT has also been studied in combination with beam radiotherapy modalities such as external beam radiation therapy and carbon ion radiation therapy to enhance the anti-tumor response. Nanotechnology aids in CRT by bringing together multiple monotherapies on a single nanostructure platform for treating cancers in a more precise or personalized way. CRT will be a key player in managing cancers if correctly tailored to the individual patient profile. The success of CRT lies in an in-depth understanding of the radiobiological principles and pathways activated in response.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Suresh Subramanian
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre; Homi Bhabha National Institute, Mumbai 400094, India.
| |
Collapse
|
19
|
Fishbein L, Del Rivero J, Else T, Howe JR, Asa SL, Cohen DL, Dahia PLM, Fraker DL, Goodman KA, Hope TA, Kunz PL, Perez K, Perrier ND, Pryma DA, Ryder M, Sasson AR, Soulen MC, Jimenez C. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Management of Metastatic and/or Unresectable Pheochromocytoma and Paraganglioma. Pancreas 2021; 50:469-493. [PMID: 33939658 DOI: 10.1097/mpa.0000000000001792] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT This manuscript is the result of the North American Neuroendocrine Tumor Society consensus conference on the medical management and surveillance of metastatic and unresectable pheochromocytoma and paraganglioma held on October 2 and 3, 2019. The panelists consisted of endocrinologists, medical oncologists, surgeons, radiologists/nuclear medicine physicians, nephrologists, pathologists, and radiation oncologists. The panelists performed a literature review on a series of questions regarding the medical management of metastatic and unresectable pheochromocytoma and paraganglioma as well as questions regarding surveillance after resection. The panelists voted on controversial topics, and final recommendations were sent to all panel members for final approval.
Collapse
Affiliation(s)
- Lauren Fishbein
- From the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tobias Else
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - James R Howe
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Sylvia L Asa
- Department of Pathology, University Hospitals Cleveland Medical Center and University Health Network, Toronto, Case Western Reserve University, Cleveland, OH
| | - Debbie L Cohen
- Renal Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Patricia L M Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX
| | - Douglas L Fraker
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania and Abramson Cancer Center, Philadelphia, PA
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA
| | - Pamela L Kunz
- Division of Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nancy D Perrier
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Daniel A Pryma
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mabel Ryder
- Endocrine Oncology Tumor Group, Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | - Aaron R Sasson
- Division of Surgical Oncology, Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY
| | - Michael C Soulen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Barnabas R, Jaiswal SK, Memon SS, Sarathi V, Malhotra G, Verma P, Patil VA, Lila AR, Shah NS, Bandgar TR. Low-Dose, Low-Specific Activity 131I-metaiodobenzyl Guanidine Therapy in Metastatic Pheochromocytoma/Sympathetic Paraganglioma: Single-Center Experience from Western India. Indian J Endocrinol Metab 2021; 25:148-159. [PMID: 34660244 PMCID: PMC8477742 DOI: 10.4103/ijem.ijem_52_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Radionuclide therapy is a promising treatment modality in metastatic pheochromocytoma/paraganglioma (PPGL). There is scarce data on 131I-metaiodobenzyl guanidine (131I-MIBG) therapy from the Indian subcontinent. Hence, we aim to study the safety and effectiveness of low-dose, low-specific activity (LSA) 131I-MIBG therapy in patients with symptomatic, metastatic PPGL. METHODS Clinical, hormonal, and radiological response parameters and side effects of LSA 131I-MIBG therapy in patients with symptomatic, metastatic PPGL were retrospectively reviewed. World health organizations' (WHO) symptomatic, hormonal, and tumor response, and response evaluation criteria in solid tumors (RECIST1.1) criteria were used to assess the response. RESULTS Seventeen (PCC: 11, sympathetic PGL: 06) patients (15 with disease progression) received low-dose LSA 131I-MIBG therapy. Complete remission (CR), partial remission (PR), stable disease (SD), and progressive disease (PD) were 18% (3/17), 24% (4/17), 18% (3/17), and 41% (7/17), respectively, for WHO symptomatic response; 20% (2/10), 10% (1/10), 30% (3/10), and 40% (4/10), respectively, for WHO hormonal response; and 19% (3/16), 6% (1/16), 31% (5/16), and 44% (7/16), respectively for tumor response based on RECIST1.1. All patients with symptomatic PD and 50% (2/4) with hormonal PD had progression as per RECIST1.1 criteria. Side effects included thrombocytopenia, acute myeloid leukemia, mucoepidermoid carcinoma, and azoospermia in 6% (1/17) each. CONCLUSIONS Our study reaffirms the modest efficacy and safety of low-dose, LSA 131I-MIBG therapy in patients with symptomatic, metastatic PPGL. Symptomatic, but not hormonal, progression after 131I-MIBG therapy correlates well with tumor progression and should be further evaluated with imaging. In resource-limited settings, anatomic imaging alone may be used to assess tumor response to 131I-MIBG therapy.
Collapse
Affiliation(s)
- Rohit Barnabas
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Sanjeet Kumar Jaiswal
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Saba Samad Memon
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Center, Bengaluru, Karnataka, India
| | - Gaurav Malhotra
- Department of Nuclear Medicine, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| | - Priyanka Verma
- Department of Nuclear Medicine, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| | - Virendra A. Patil
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Anurag R. Lila
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Nalini S. Shah
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Tushar R. Bandgar
- Department of Endocrinology, Seth G.S Medical College and KEM Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
21
|
Grambozov B, Sedlmayer F, Lottspeich C, Reincke M, Zehentmayr F. Vertebral metastases of functional vaginal paraganglioma treated with radiation therapy: A case report. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2020. [DOI: 10.1016/j.cpccr.2020.100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Pediatric pheochromocytoma: current status of diagnostic imaging and treatment procedures. Curr Opin Urol 2020; 29:493-499. [PMID: 31246590 DOI: 10.1097/mou.0000000000000650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To provide an overview of relevant data available and updated recommendations for management of pediatric patients with pheochromocytoma (PCC). RECENT FINDINGS Much of the available data surrounding pediatric PCC is in the form of case reports and case series. With the accumulation of data over time, pediatric PCC does in fact differ significantly from not only what is known in the adult population, but also from classic teaching. Pediatric patients are much more likely to have a hereditary predisposition as well as aggressive and malignant disease. Much of the recent literature focuses on defining these genetic syndromes in order to provide recommendations for screening and genetic counseling. Other recent advances center around developing treatments for metastatic disease. Timely diagnosis with plasma metanephrines and cross-sectional imaging, and appropriate preoperative medical optimization followed by surgical resection remain the center of treatment. SUMMARY Although rare and adult principles are applied to pediatric PCC, genetic testing plays a pivotal role in management of children, adolescents and young adults with PCC.
Collapse
|
23
|
Jasim S, Jimenez C. Metastatic pheochromocytoma and paraganglioma: Management of endocrine manifestations, surgery and ablative procedures, and systemic therapies. Best Pract Res Clin Endocrinol Metab 2020; 34:101354. [PMID: 31685417 DOI: 10.1016/j.beem.2019.101354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Metastatic pheochromocytomas and paragangliomas (MPPGs) are rare neuroendocrine tumors. Most patients present with advanced disease that is associated with manifestations of catecholamine release. Surgical resection of the primary tumor and ablative therapies of metastases-whenever possible-may improve clinical outcomes and, perhaps, lengthen the patient's overall survival. Significant steps in understanding the genetic alterations linked to MPPGs and scientific progress made on cancers that share a similar pathogenesis are leading to the recognition of potential systemic therapeutic options. Data derived from clinical trials evaluating targeted therapies such as tyrosine kinase inhibitors, radiopharmaceuticals, immunotherapy, and combinations of these will likely improve the outcomes of patients with advanced and progressive MPPGs. Exemplary of this success is the recent approval in the United States of the high-specific-activity iodine131 meta-iodine-benzylguanidine (MIBG) for patients with unresectable and progressive MPPGs that express the noradrenaline transporter. This review will discuss the therapeutic approaches for patients with MPPGs.
Collapse
Affiliation(s)
- Sina Jasim
- The Division of Endocrinology, Metabolism and Lipid Research, Campus box 8127, Washington University, School of Medicine, 660 S. Euclid Ave., St. Louis, Missouri 63110, USA
| | - Camilo Jimenez
- The Department of Endocrine Neoplasia and Hormonal Disorders at the University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1461, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Rocco R, Murphy BL, Patel VP, Hamidi O, Chang AY, Lyden ML. A rare case of a 65 year old female with a mesenteric paraganglioma. HUMAN PATHOLOGY: CASE REPORTS 2020. [DOI: 10.1016/j.ehpc.2019.200349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
25
|
Jimenez C, Erwin W, Chasen B. Targeted Radionuclide Therapy for Patients with Metastatic Pheochromocytoma and Paraganglioma: From Low-Specific-Activity to High-Specific-Activity Iodine-131 Metaiodobenzylguanidine. Cancers (Basel) 2019; 11:cancers11071018. [PMID: 31330766 PMCID: PMC6678905 DOI: 10.3390/cancers11071018] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/03/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Low-specific-activity iodine-131–radiolabeled metaiodobenzylguanidine (I-131-MIBG) was introduced last century as a potential systemic therapy for patients with malignant pheochromocytomas and paragangliomas. Collective information derived from mainly retrospective studies has suggested that 30–40% of patients with these tumors benefit from this treatment. A low index of radioactivity, lack of therapeutic standardization, and toxicity associated with intermediate to high activities (absorbed radiation doses) has prevented the implementation of I-131-MIBG’s in clinical practice. High-specific-activity, carrier-free I-131-MIBG has been developed over the past two decades as a novel therapy for patients with metastatic pheochromocytomas and paragangliomas that express the norepinephrine transporter. This drug allows for a high level of radioactivity, and as yet is not associated with cardiovascular toxicity. In a pivotal phase two clinical trial, more than 90% of patients achieved partial responses and disease stabilization with the improvement of hypertension. Furthermore, many patients exhibited long-term persistent antineoplastic effects. Currently, the high-specific-activity I-131-MIBG is the only approved therapy in the US for patients with metastatic pheochromocytomas and paragangliomas. This review will discuss the historical development of high-specific-activity I-131-MIBG, its benefits and adverse events, and future directions for clinical practice applicability and trial development.
Collapse
Affiliation(s)
- Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1461, Houston, TX 77030, USA.
| | - William Erwin
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1461, Houston, TX 77030, USA
| | - Beth Chasen
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
26
|
Afani L, Errihani H, Awada A. [Therapeutic management of malignant paragangliomas: an illustrative case and literature review]. Pan Afr Med J 2019; 32:62. [PMID: 31223354 PMCID: PMC6560965 DOI: 10.11604/pamj.2019.32.62.8812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/31/2016] [Indexed: 11/28/2022] Open
Abstract
Les paragangliomes malins présentent un réel challenge pour les praticiens. Il s'agit de tumeurs rares, de caractérisation complexe, et très hétérogènes dans leur évolution et pronostic. Devant la rareté de ce groupe tumoral, il n'existe pas de consensus sur la prise en charge thérapeutique. A travers ce cas illustratif, nous rapportons l'observation d'une patiente de 29 ans suivie pour un paragangliome malin traitée initialement par chirurgie. Après six mois d'intervalle, récidive multifocale fixant à l'Octreo PET-CT (Positron Emission Tomography- Computer Tomography), la patiente a été mise sous injection mensuelle de Somatuline pendant un an puis arrêté suite à une progression. Une 2ème chirurgie de cytoréduction a été réalisée suivie d'une radiothérapie. Après un an, la patiente a présenté une progression massive. Une chimiothérapie à base de Dacarbazine a été initiée et dont l'évaluation a montré une réponse métabolique quasi complète après huit cures. Le but de ce travail est d'exposer les différentes options thérapeutiques possibles dans la prise en charge des paragangliomes malins.
Collapse
Affiliation(s)
- Leila Afani
- Service d'Oncologie Médicale, Institut National d'Oncologie, Rabat, Maroc
| | - Hassan Errihani
- Service d'Oncologie Médicale, Institut National d'Oncologie, Rabat, Maroc
| | - Ahmad Awada
- Service d'Oncologie Médicale, Institut Jules Bordet, Bd de Waterloo, Bruxelles, Belgique
| |
Collapse
|
27
|
Mesko S, Deegan BJ, D'Souza NM, Ghia AJ, Chapman BV, Amini B, McAleer MF, Wang XA, Brown PD, Tatsui CE, Rhines L, Li J. Spine Stereotactic Radiosurgery for Metastatic Pheochromocytoma. Cureus 2019; 11:e4742. [PMID: 31355101 PMCID: PMC6649891 DOI: 10.7759/cureus.4742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Purpose: Despite aggressive primary treatment, up to 13.5% of patients diagnosed with pheochromocytoma may develop metastases, most often affecting the axial skeleton. Given that systemic therapy options are often inadequate, local therapy remains the cornerstone of palliation for these patients. Historically poor responses to standard fractionated radiotherapy have led to the consideration of stereotactic radiosurgery as an option to overcome potential radioresistance and provide durable local control of these tumors. Here we report our institutional experience in treating spine metastases from pheochromocytoma with spine stereotactic radiosurgery (SSRS). Methods and materials: Our clinical databases were retrospectively reviewed for patients with metastatic pheochromocytoma treated with SSRS from 2000-2017. Seven patients with 16 treated metastatic spinal lesions were identified. Local control was evaluated using magnetic resonance imaging (MRI). Pain and symptom data were assessed to evaluate toxicity using Common Terminology Criteria for Adverse Events (CTCAE) v4.03. The Kaplan-Meier method was used to assess local control and overall survival (OS). Results: Median follow-up for treated lesions was 11 months (range 2.2 - 70.8). Most lesions were treated to a dose of 27 Gy in three fractions (62.5%). Other fractionation schemes included 24 Gy in one fraction (25%), 16 Gy in one fraction (6.3%), and 18 Gy in three fractions (6.3%). Treatment sites included the cervical spine (18.8%), thoracic spine (37.5%), lumbar spine (31.3%), and sacrum (12.5%). The crude local control rate was 93.7%, with one thoracic spine lesion progressing 20.7 months after treatment with 24 Gy in one fraction. Kaplan-Meier OS rates at 1 and 2 years after SSRS were 71.4% and 42.9%, respectively. Most common toxicities included acute grade 1-2 pain and fatigue. There was one case of vertebral fracture in a cervical spine lesion treated to 27 Gy in three fractions, which was managed non-surgically. Conclusion: Very few studies have explored the use of SSRS in metastatic pheochromocytoma. Our data suggest this modern radiation modality is effective, safe, and provides durable local control to palliate symptoms and potentially limit further metastatic seeding. Larger patient numbers and longer follow-up will further define the role of SSRS as a treatment option in these patients.
Collapse
Affiliation(s)
- Shane Mesko
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Brian J Deegan
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Neil M D'Souza
- Radiation Oncology, Mays Cancer Center, University of Texas, San Antonio, USA
| | - Amol J Ghia
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Bhavana V Chapman
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Behrang Amini
- Radiology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Mary Frances McAleer
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Xin A Wang
- Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | | | - Claudio E Tatsui
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Laurence Rhines
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jing Li
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
28
|
Yadav MP, Ballal S, Bal C. Concomitant 177Lu-DOTATATE and capecitabine therapy in malignant paragangliomas. EJNMMI Res 2019; 9:13. [PMID: 30725219 PMCID: PMC6365580 DOI: 10.1186/s13550-019-0484-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The role of concomitant peptide receptor radionuclide therapy (PRRT) and capecitabine therapy has shown benefit in gastroenteropancreatic neuroendocrine tumors. However, data reporting its role in paraganglioma (PGL) patients is lacking. The aim of this study was to evaluate the role of combined capecitabine and 177Lu-DOTATATE in malignant PGL patients. METHODS In this retrospective, single-institutional, single-arm, observational study, data of consecutive advanced stage PGL patients treated with concomitant 177Lu-DOTATATE-capecitabine therapy, between July 2009 and March 2017, were collected and analyzed. RESULTS Twenty-five PGL patients received an average dose of 22.86 ± 9.54 (14.43-50) GBq 177Lu-DOTATATE and 1250 mg/m2 capecitabine from days 0 to 14, commencing on the morning of PRRT. The median overall survival (OS) was not attained in this patient cohort; however, the median PFS was 32 months. Morphological response according to RECIST 1.1 criteria was achieved in 28% (7/25) patients. Biochemical response with > 50% reduction in chromogranin A levels was observed in 28% of the patients. CONCLUSIONS Our data confirm that 177Lu-DOTATATE-capecitabine therapy is effective in achieving an objective response in 28% and symptomatic response in 43% patients. In comparison to published PRRT monotherapy outcomes in PGL, we did not observe any great advantage of concomitant therapy; however, it could be due to under-powered study. We recommend a large randomized trial to prove or disprove the utility of capecitabine as a radiosensitizer for PRRT in PGL patients.
Collapse
Affiliation(s)
- Madhav Prasad Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sanjana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Pheochromocytomas and paragangliomas (PPGLs) are uncommon catecholamine-producing neuroendocrine neoplasms that usually present with secondary hypertension. This review is to update the current knowledge about these neoplasms, the pathophysiology, genetic aspects and diagnostic and therapeutic algorithms based on scientific literature mostly within the past 3 years. RECENT FINDINGS Eighty to eighty-five percent of PPGLs arise from the adrenal medulla (pheochromocytomas; PCCs) and the remainder from the autonomic neural ganglia (paragangliomas; PGLs). Catecholamine excess causes chronic or paroxysmal hypertension associated with sweating, headaches and palpitations, the presenting features of PPGLs, and increases the cardiovascular morbidity and mortality. Genetic testing should be considered in all cases as mutations are reported in 35-40% of cases; 10-15% of PCCs and 20-50% of PGLs can be malignant. Measurements of plasma-free metanephrines or 24-h urine-fractionated metanephrines help biochemical diagnosis with high sensitivity and specificity. Initial anatomical localization after biochemical confirmation is usually with computed tomography (CT) or magnetic resonance imaging (MRI). 123Iodine metaiodobenzylguanidine (123I-MIBG) scintigraphy, positron emission tomography (PET) or single-photon emission computed tomography (SPECT) is often performed for functional imaging and prognostication prior to curative or palliative surgery. Clinical and biochemical follow-up is recommended at least annually after complete tumour excision. Children, pregnant women and older people have higher morbidity and mortality risk. De-bulking surgery, chemotherapy, radiotherapy, radionuclide agents and ablation procedures are useful in the palliation of incurable disease. PPGLs are unique neuroendocrine tumours that form an important cause for endocrine hypertension. The diagnostic and therapeutic algorithms are updated in this comprehensive article.
Collapse
Affiliation(s)
- Joseph M Pappachan
- Department of Endocrinology and Metabolism, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, LA1 4RP, UK.
| | - Nyo Nyo Tun
- Metabolic Unit, Western General Hospital, Edinburgh, UK
| | | | - Ravinder Sodi
- Department of Biochemistry and Blood Sciences, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, LA1 4RP, UK
| | - Fahmy W F Hanna
- Department of Endocrinology and Metabolism, The Royal Stoke University Hospital and North Staffordshire University, Stoke-on-Trent, ST4 6QG, UK
| |
Collapse
|
30
|
Breen W, Bancos I, Young WF, Bible KC, Laack NN, Foote RL, Hallemeier CL. External beam radiation therapy for advanced/unresectable malignant paraganglioma and pheochromocytoma. Adv Radiat Oncol 2017; 3:25-29. [PMID: 29556576 PMCID: PMC5856976 DOI: 10.1016/j.adro.2017.11.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/26/2017] [Accepted: 11/15/2017] [Indexed: 11/18/2022] Open
Abstract
Purpose/Objectives To evaluate the role of external beam radiation therapy (EBRT) for treatment of malignant paraganglioma (PGL) and pheochromocytoma (PCC). Methods and materials A retrospective review was performed of all patients with malignant PGL/PCC treated with EBRT at our institution between 1973 and 2015. Local control (LC) per treated lesion and overall survival were estimated using the Kaplan-Meier method. Toxicities were scored using the Common Toxicity Criteria for Adverse Events (AE), version 4. Results The cohort included 41 patients with 107 sites treated. Median (range) age at EBRT was 33 (11-80) years. Treatment intention was curative in 20 patients (30 lesions) and palliative in 21 patients (77 lesions). The primary tumor was PGL (63%) and PCC (37%). Previous local therapies were surgical resection (90%) and percutaneous ablation (19%). Indications for EBRT were local control (66%), pain (22%), or spinal cord compression (12%). Treatment site included bone (69%), soft tissue (30%), and liver (1%). Median (range) EBRT dose was 40 (6.5-70) Gy. Median biologic effective dose using α/β = 10 (BED10) was 53 (9-132). Median follow-up was 3.8 years (0.04-41.5), and mean follow-up was 9.7 years. Overall survival at 5 years was 65%: 79% for curative- and 50% for palliative-intention patients (P = .028). LC at 5 years was 81% for all lesions; 91% for lesions receiving BED10 ≥53, and 62% for lesions receiving BED10 <53 (P = .001). All 11 lesions treated with stereotactic body RT or radiosurgery had LC at a median of 3.0 (0.2-5.4) years. For the symptomatic lesions, symptoms improved in 94%. There were no acute grade ≥3 treatment-related AEs, including no hypertensive crises. Two patients developed a late grade ≥3 AE. Conclusions EBRT is a useful treatment modality for malignant PGL and PCC. Higher RT dose was associated with improved LC.
Collapse
Affiliation(s)
- William Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Irina Bancos
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Keith C. Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Nadia N. Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Robert L. Foote
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Christopher L. Hallemeier
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
- Corresponding author. Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905.Department of Radiation OncologyMayo Clinic200 First Street SWRochesterMN55905
| |
Collapse
|
31
|
Corroyer-Dulmont A, Falzone N, Kersemans V, Thompson J, Allen DP, Able S, Kartsonaki C, Malcolm J, Kinchesh P, Hill MA, Vojnovic B, Smart SC, Gaze MN, Vallis KA. Improved outcome of 131I-mIBG treatment through combination with external beam radiotherapy in the SK-N-SH mouse model of neuroblastoma. Radiother Oncol 2017; 124:488-495. [PMID: 28595752 PMCID: PMC5636618 DOI: 10.1016/j.radonc.2017.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/03/2022]
Abstract
PURPOSE To assess the efficacy of different schedules for combining external beam radiotherapy (EBRT) with molecular radiotherapy (MRT) using 131I-mIBG in the management of neuroblastoma. MATERIALS AND METHODS BALB/c nu/nu mice bearing SK-N-SH neuroblastoma xenografts were assigned to five treatment groups: 131I-mIBG 24h after EBRT, EBRT 6days after 131I-mIBG, EBRT alone, 131I-mIBG alone and control (untreated). A total of 56 mice were assigned to 3 studies. Study 1: Vessel permeability was evaluated using dynamic contrast-enhanced (DCE)-MRI (n=3). Study 2: Tumour uptake of 131I-mIBG in excised lesions was evaluated by γ-counting and autoradiography (n=28). Study 3: Tumour volume was assessed by longitudinal MR imaging and survival was analysed (n=25). Tumour dosimetry was performed using Monte Carlo simulations of absorbed fractions with the radiation transport code PENELOPE. RESULTS Given alone, both 131I-mIBG and EBRT resulted in a seven-day delay in tumour regrowth. Following EBRT, vessel permeability was evaluated by DCE-MRI and showed an increase at 24h post irradiation that correlated with an increase in 131I-mIBG tumour uptake, absorbed dose and overall survival in the case of combined treatment. Similarly, EBRT administered seven days after MRT to coincide with tumour regrowth, significantly decreased the tumour volume and increased overall survival. CONCLUSIONS This study demonstrates that combining EBRT and MRT has an enhanced therapeutic effect and emphasizes the importance of treatment scheduling according to pathophysiological criteria such as tumour vessel permeability and tumour growth kinetics.
Collapse
Affiliation(s)
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Veerle Kersemans
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - James Thompson
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Danny P Allen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Sarah Able
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | | | - Javian Malcolm
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Paul Kinchesh
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Mark A Hill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Boris Vojnovic
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Sean C Smart
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK
| | - Mark N Gaze
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, Oxford University, UK.
| |
Collapse
|
32
|
Fishbein L, Ben-Maimon S, Keefe S, Cengel K, Pryma DA, Loaiza-Bonilla A, Fraker DL, Nathanson KL, Cohen DL. SDHB mutation carriers with malignant pheochromocytoma respond better to CVD. Endocr Relat Cancer 2017; 24:L51-L55. [PMID: 28566531 DOI: 10.1530/erc-17-0086] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/31/2017] [Indexed: 11/08/2022]
Affiliation(s)
- L Fishbein
- Division of EndocrinologyMetabolism and Diabetes and Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - S Ben-Maimon
- Department of MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Renal and Hypertension DivisionPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - S Keefe
- Department of MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Medical Oncology and HematologyPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - K Cengel
- Department of Radiation OncologyPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - D A Pryma
- Department of RadiologyPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Nuclear MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - A Loaiza-Bonilla
- Department of MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Medical Oncology and HematologyPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - D L Fraker
- Department of SurgeryPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Endocrine Oncologic SurgeryPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - K L Nathanson
- Department of MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Translational Medicine and Human GeneticsPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer CenterUniversity of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - D L Cohen
- Department of MedicinePerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Renal and Hypertension DivisionPerelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Bholah R, Bunchman TE. Review of Pediatric Pheochromocytoma and Paraganglioma. Front Pediatr 2017; 5:155. [PMID: 28752085 PMCID: PMC5508015 DOI: 10.3389/fped.2017.00155] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/26/2017] [Indexed: 01/01/2023] Open
Abstract
Pheochromocytoma (PCC) and paraganglioma (PGL) are rare chromaffin cell tumors which secrete catecholamines and form part of the family of neuroendocrine tumors. Although a rare cause of secondary hypertension in pediatrics, the presentation of hypertension in these patients is characteristic, and treatment is definitive. The gold standard for diagnosis is via measurement of plasma free metanephrines, with imaging studies performed for localization, identification of metastatic lesions and for surgical resection. Preoperative therapy with alpha-blocking agents, beta blockers, and potentially tyrosine hydroxylase inhibitors aid in a safe pre-, intra- and postoperative course. PCC and PGL are inherited in as much as 80% of pediatric cases, and all patients with mutations should be followed closely given the risk of recurrence and malignancy. While the presentation of chromaffin cell tumors has been well described with multiple endocrine neoplasia, NF1, and Von Hippel-Lindau syndromes, the identification of new gene mutations leading to chromaffin cell tumors at a young age is changing the landscape of how clinicians approach such cases. The paraganglioma-pheochromocytoma syndromes (SDHx) comprise familial gene mutations, of which the SDHB gene mutation carries a high rate of malignancy. Since the inheritance rate of such tumors is higher than previously described, genetic screening is recommended in all patients, and lifelong follow-up for recurrent tumors is a must. A multidisciplinary team approach allows for optimal health-care delivery in such children. This review serves to provide an overview of pediatric PCC and PGL, including updates on the preferred methods of imaging, guidelines on gene testing as well as management of hypertension in such patients.
Collapse
Affiliation(s)
- Reshma Bholah
- Pediatric Nephrology, Virginia Commonwealth University, Richmond, VA, United States
| | | |
Collapse
|
34
|
Gill MR, Falzone N, Du Y, Vallis KA. Targeted radionuclide therapy in combined-modality regimens. Lancet Oncol 2017; 18:e414-e423. [PMID: 28677577 DOI: 10.1016/s1470-2045(17)30379-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Targeted radionuclide therapy (TRT) is a branch of cancer medicine concerned with the use of radioisotopes, radiolabelled molecules, nanoparticles, or microparticles that either naturally accumulate in or are designed to target tumours. TRT combines the specificity of molecular and sometimes physical targeting with the potent cytotoxicity of ionising radiation. Targeting vectors for TRT include antibodies, antibody fragments, proteins, peptides, and small molecules. The diversity of available carrier molecules, together with the large panel of suitable radioisotopes with unique physicochemical properties, allows vector-radionuclide pairings to be matched to the molecular, pathological, and physical characteristics of a tumour. Some pairings are designed for dual therapeutic and diagnostic applications. Use of TRT is increasing with the adoption into practice of radium-223 dichloride for the treatment of bone metastases and with the ongoing clinical development of, among others, 177Lu-dodecanetetraacetic acid tyrosine-3-octreotate (DOTATATE) for the treatment of neuroendocrine tumours and 90Y-microspheres for the treatment of hepatic tumours. The increasing use of TRT raises the question of how best to integrate TRT into multimodality protocols. Achievements in this area and the future prospects of TRT are evaluated in this Review.
Collapse
Affiliation(s)
- Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yong Du
- The Royal Marsden Hospital NHS Foundation Trust, Sutton, Surrey, UK
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Prades CA, Atassi B, Nazeer H. Metastatic Malignant Paraganglioma: A Case Report and Review of Literature. World J Oncol 2017; 8:92-95. [PMID: 29147442 PMCID: PMC5650004 DOI: 10.14740/wjon1033w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2017] [Indexed: 11/11/2022] Open
Abstract
Metastasis is a rare presentation of non-secretory paraganglioma. Consequently, there is no standard of care for the treatment of metastatic malignant paraganglioma. The most widely used chemotherapy regimen for non-resectable cases includes cyclophosphamide, vincristine, and dacarbazine (CVD). CVD has been previously studied with variable therapeutic response. However, yttrium-90 (Y90) radioembolization has not been previously studied in the treatment of hepatic metastasis in patients diagnosed with malignant paraganglioma. This case report follows the treatment of a patient with stage IV mediastinal paraganglioma with metastasis to the liver. Treatment consisted of the CVD chemotherapy regimen and Y90 radioembolization of the hepatic lesions. After 10 cycles of CVD, the tumor size has decreased from 6.0 × 8.8 to 5.5 × 3.0 × 3.4 cm on computed tomography scan. The prominent metastatic liver lesions responded after Y90 radioembolization of both the right and left hepatic arteries. The prominent right hepatic lobe lesion has decreased in size from 2.6 × 3.4 × 3.0 cm to 2.6 × 2.8 × 2.9 cm. The prominent left hepatic lobe lesion originally measuring 1.6 cm in diameter completely resolved on follow-up imaging studies. After completion of 12 cycles of chemotherapy, the most recent positron emission testing scan determined no evidence of disease regarding both the primary mass and the hepatic lesions. This study demonstrates the first case of combination chemotherapy and Y90 radioembolization with a complete response per response evaluation criteria in solid tumors criteria. The approaches toward diagnosis and treatment corresponding to this case of malignant metastatic paraganglioma are also reviewed in this study.
Collapse
Affiliation(s)
| | - Bassel Atassi
- Chicago Medical School, Woodridge, IL, USA.,Cancer Center, Little Company of Mary Hospital, Evergreen Park, IL, USA
| | - Hamid Nazeer
- Little Company of Mary Hospital, Evergreen Park, IL, USA
| |
Collapse
|
36
|
Chaaya G, Morales J, Castiglioni A, Subhani N, Asmar A. Paraganglioma of the Urinary Bladder: A Rare Cause of Hypertension and Urinary Tract Infections. Am J Med Sci 2017; 355:191-194. [PMID: 29406048 DOI: 10.1016/j.amjms.2017.03.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/28/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Pheochromocytoma is a neoplasm, which develops from cells of the chromaffin tissues that are derived from the ectodermic neural system and mostly situated within the adrenal medulla. Approximately 15% of pheochromocytoma cases arise from extra-adrenal chromaffin tissue. Pheochromocytoma of the bladder is rare and accounts for less than 0.06% of all bladder neoplasms and less than 1% of all pheochromocytomas. We report a case of a young woman who presented with uncontrolled hypertension, recurrent urinary tract infections and micturition attacks and was found to have a metastatic bladder paraganglioma. In addition, we provide a summary table of the clinical manifestations of paragangliomas based on anatomic locations.
Collapse
Affiliation(s)
- Gerard Chaaya
- University of Central Florida College of Medicine, Orlando, Florida
| | - Jorge Morales
- University of Central Florida College of Medicine, Orlando, Florida
| | - Analia Castiglioni
- University of Central Florida College of Medicine, Orlando, Florida; Orlando VA Medical Center, Orlando, Florida
| | - Noman Subhani
- University of Central Florida College of Medicine, Orlando, Florida; Osceola Regional Medical Center, Kissimmee, Florida
| | - Abdo Asmar
- University of Central Florida College of Medicine, Orlando, Florida; Orlando VA Medical Center, Orlando, Florida.
| |
Collapse
|
37
|
Thomson N, Pacak K, Schmidt M, Palmer C, Salzman K, Champine M, Schiffman J, Cohen A. Leptomeningeal dissemination of a low-grade lumbar paraganglioma: case report. J Neurosurg Spine 2017; 26:501-506. [PMID: 28128698 DOI: 10.3171/2016.10.spine16948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Leptomeningeal dissemination of paraganglioma is rare, with only 2 prior cases in the literature. The authors present the case of a metastatic low-grade lumbar paraganglioma via leptomeningeal dissemination. This report emphasizes the utility of 3,4-dihydroxy-6-18F-fluoro-l-phenylalanine (18F-FDOPA) PET scanning for diagnosis, as well as the combination of radiation therapy and alkylating chemotherapeutic agents for the treatment of this rare phenomenon. The patient was a 61-year-old woman who presented with low-back pain and was found to have an isolated L-3 intrathecal tumor on MRI. Sixteen months after gross-total en bloc resection of the paraganglioma, the patient again became symptomatic with new neurological symptoms. MRI findings revealed enhancing leptomeningeal nodules throughout the spine. 18F-FDOPA PET/CT scanning was used to confirm the diagnosis of disseminated paraganglioma. Intrathecal thiotepa, radiation therapy, and systemic therapy with capecitabine and temozolomide have been used sequentially over a 2-year period, with each able to stabilize tumor growth for several months. The authors also summarize the 2 other reports of leptomeningeal dissemination of paragangliomas in the literature and compare the course and management of the 3 cases.
Collapse
Affiliation(s)
- Nick Thomson
- Department of Internal Medicine, University of Arizona College of Medicine Phoenix, Arizona
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Meic Schmidt
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Cheryl Palmer
- Department of Pathology, Division of Anatomic Pathology, University of Utah, Salt Lake City, Utah,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Karen Salzman
- Department of Radiology, Division of Neuroradiology, University of Utah, Salt Lake City, Utah,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Marjan Champine
- Huntsman Cancer Institute, Salt Lake City, Utah,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Joshua Schiffman
- Department of Pediatrics and Oncological Sciences, University of Utah, Salt Lake City, Utah,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Adam Cohen
- Department of Medicine, Division of Oncology, University of Utah, Salt Lake City, Utah,Huntsman Cancer Institute, Salt Lake City, Utah
| |
Collapse
|
38
|
Mittal J, Manikandan R, Dorairajan LN, Toi PC. Recurrent Malignant Pheochromocytoma with Lymph Nodal Metastasis in a Child: A Rare Case. J Indian Assoc Pediatr Surg 2017; 22:242-244. [PMID: 28974878 PMCID: PMC5615900 DOI: 10.4103/0971-9261.214454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Malignant pheochromocytoma in children manifesting as local recurrence with multiple lymph nodal metastasis is a rare entity. We report a case of a 14-year-old child with recurrent sporadic malignant pheochromocytoma presenting 8 years after primary surgery with retroperitoneal lymphadenopathy. The child underwent excision of the tumor along with retroperitoneal lymphadenectomy. Histopathology confirmed pheochromocytoma with extensive lymph node metastasis. The patient has no signs of disease recurrence till date. This report supports the long-term follow-up and aggressive surgical approach to remove all foci of tumor in recurrent pheochromocytoma.
Collapse
Affiliation(s)
| | | | | | - Pampa Ch Toi
- Department of Pathology, JIPMER, Puducherry, India
| |
Collapse
|
39
|
Violante AHD, Ortiga-Carvalho TM, Soares da Costa MH, Vaisman M. Treatment of pheochromocytomas and paragangliomas: genetic approach? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2016-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pheochromocytomas (PHEOs) and paragangliomas (PGLs) are neuroendocrine tumors derived from the chromaffin tissue of the adrenal medulla or from extra-adrenal sympathetic and parasympathetic paraganglia. These tumors affect one in 2500–6500 people, with 500–1600 patients diagnosed annually in the USA. Its real incidence is likely to be much higher, and many patients may die undiagnosed. It is a rare cause of secondary hypertension, with an incidence of 0.1–0.6%. PHEOs are more common than PGLs, and 5–7% is related to hormonal secretion. The average age of diagnosis is 43 years, but 10–20% of PHEO/PGL patients are children, and the tumors are generally associated with genetic conditions. The worldwide consensus is that people with PHEOs/PGLs should undergo a mandatory genetic investigation, especially with multiple injuries and those who are younger. Although many PHEOs are sporadic and benign, approximately 30% of familial tumor cases have been identified. Multiple endocrine neoplasia type 2, von Hippel–Lindau syndrome and neurofibromatosis type 1 are classically linked to PHEO and mutations in succinate dehydrogenase complex subunits have been associated with familial forms. Our goal is to review various clinical syndromes in which PHEOs/PGLs are present, along with their therapeutic options.
Collapse
Affiliation(s)
| | - Tania M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Mario Vaisman
- Division of Endocrinology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare but unique neuroendocrine tumors. The hypersecretion of catecholamines from the tumors can be associated with high morbidity and mortality, even when tumors are benign. Up to 40% of PCCs/PGLs are associated with germline mutations in susceptibility genes. About one-quarter are malignant, defined by the presence of distant metastases. Treatment options for unresectable metastatic disease, including chemotherapy, (131)I-MIBG, and radiation, can offer limited tumor and hormone control, although none are curative. This article reviews the inherited genetics, diagnosis, and treatment of PCCs and PGLs.
Collapse
Affiliation(s)
- Lauren Fishbein
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 351 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Persky MJ, Adelman M, Zias E, Myssiorek D. Necessity for lifelong follow-up of patients with familial paraganglioma syndrome: A case report. Head Neck 2015; 37:E174-8. [DOI: 10.1002/hed.24047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2015] [Indexed: 01/21/2023] Open
Affiliation(s)
- Michael J. Persky
- Department of Otolaryngology - Head and Neck Surgery; New York University Langone Medical Center, Clinical Cancer Center; New York New York
| | - Mark Adelman
- Department of Vascular Surgery; New York University Langone Medical Center, Clinical Cancer Center; New York New York
| | - Elias Zias
- Department of Cardiovascular Surgery; New York University Langone Medical Center, Clinical Cancer Center; New York New York
| | - David Myssiorek
- Department of Otolaryngology - Head and Neck Surgery; New York University Langone Medical Center, Clinical Cancer Center; New York New York
| |
Collapse
|
42
|
Dietrich A, Koi L, Zöphel K, Sihver W, Kotzerke J, Baumann M, Krause M. Improving external beam radiotherapy by combination with internal irradiation. Br J Radiol 2015; 88:20150042. [PMID: 25782328 DOI: 10.1259/bjr.20150042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of external beam radiotherapy (EBRT) is dose dependent, but the dose that can be applied to solid tumour lesions is limited by the sensitivity of the surrounding tissue. The combination of EBRT with systemically applied radioimmunotherapy (RIT) is a promising approach to increase efficacy of radiotherapy. Toxicities of both treatment modalities of this combination of internal and external radiotherapy (CIERT) are not additive, as different organs at risk are in target. However, advantages of both single treatments are combined, for example, precise high dose delivery to the bulk tumour via standard EBRT, which can be increased by addition of RIT, and potential targeting of micrometastases by RIT. Eventually, theragnostic radionuclide pairs can be used to predict uptake of the radiotherapeutic drug prior to and during therapy and find individual patients who may benefit from this treatment. This review aims to highlight the outcome of pre-clinical studies on CIERT and resultant questions for translation into the clinic. Few clinical data are available until now and reasons as well as challenges for clinical implementation are discussed.
Collapse
Affiliation(s)
- A Dietrich
- 1 German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - L Koi
- 2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,3 Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - K Zöphel
- 1 German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,4 Clinic and Policlinic for Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - W Sihver
- 5 Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - J Kotzerke
- 1 German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,4 Clinic and Policlinic for Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - M Baumann
- 1 German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,3 Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,6 Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology, Dresden, Germany
| | - M Krause
- 1 German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,2 OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,3 Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,6 Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology, Dresden, Germany
| |
Collapse
|
43
|
Cremonesi M, Ferrari M, Botta F, Guerriero F, Garibaldi C, Bodei L, De Cicco C, Grana CM, Pedroli G, Orecchia R. Planning combined treatments of external beam radiation therapy and molecular radiotherapy. Cancer Biother Radiopharm 2014; 29:227-37. [PMID: 25006794 DOI: 10.1089/cbr.2014.1607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Molecular radiotherapy (MRT) with radiolabeled molecules has being constantly evolving, leading to notable results in cancer treatment. In some cases, the absorbed doses delivered to tumors by MRT are sufficient to obtain complete responses; in other cases, instead, to be effective, MRT needs to be combined with other therapeutic approaches. Recently, several studies proposed the combination of MRT with external beam radiation therapy (EBRT). Some describe the theoretical basis within radiobiological models, others report the results of clinical phase I-II studies aimed to assess the feasibility and tolerability. The latter includes the treatment of various tumors, such as meningiomas, paragangliomas, non-Hodgkin's lymphomas, bone, brain, hepatic, and breast lesions. The underlying principle of combined MRT and EBRT is the possibility of exploiting the full potential of each modality, given the different organs at risk. Target tissues can indeed receive a higher irradiation, while respecting the threshold limits of more than one critical tissue. Nevertheless, clinical trials are empirical and optimization is still a theoretical issue. This article describes the state of the art of combined MRT and EBRT regarding the rationale and the results of clinical studies, with special focus on the possibility of treatment improvement.
Collapse
Affiliation(s)
- Marta Cremonesi
- Department of Medical Imaging and Radiation Sciences, Istituto Europeo di Oncologia , Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pappachan JM, Raskauskiene D, Sriraman R, Edavalath M, Hanna FW. Diagnosis and management of pheochromocytoma: a practical guide to clinicians. Curr Hypertens Rep 2014; 16:442. [PMID: 24792093 DOI: 10.1007/s11906-014-0442-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pheochromocytomas (PCCs) are rare catecholamine producing neuroendocrine tumors. The majority of these tumors (85 %) arise from the adrenal medulla. Those arising from the extra-adrenal neural ganglia are called paragangliomas (PGLs). Paroxysmal hypertension with sweating, headaches and palpitation are the usual presenting features of PCCs/ PGLs. Gene mutations are reported in 32-79 % of cases, making genetic screening mandatory in all the cases. The malignancy rates are 10-15 % for PCCs and 20-50 % for PGLs. Measurement of plasma or 24-hour urinary fractionated metanephrines is the best biochemical diagnostic test. Computed tomography or magnetic resonance imaging has high sensitivity (90-100 %) and reasonable specificity (70-90 %) for the anatomical localization. The functionality is assessed by different radionuclide imaging modalities such as metaiodobenzylguanidine (MIBG) scintigraphy, positron emission tomography or single photon emission computed tomography. The only modality of curative treatment is tumor excision. Proper peri-operative management improves the surgical outcomes. Annual follow up with clinical and biochemical assessment is recommended in all the cases after treatment. Children, pregnant women and older people have higher morbidity and mortality risk. De-bulking surgery, chemotherapy, radiotherapy, molecular agents like sunitinib and everolimus, radionuclide agents and different ablation procedures may be useful in the palliation of inoperable/metastatic disease. An update on the diagnostic evaluation and management of PCCs and PGLs is presented here.
Collapse
Affiliation(s)
- Joseph M Pappachan
- Department of Endocrinology, Walsall Manor Hospital, West Midlands, WS2 9PS, UK,
| | | | | | | | | |
Collapse
|
45
|
Vogel J, Atanacio AS, Prodanov T, Turkbey BI, Adams K, Martucci V, Camphausen K, Fojo AT, Pacak K, Kaushal A. External beam radiation therapy in treatment of malignant pheochromocytoma and paraganglioma. Front Oncol 2014; 4:166. [PMID: 25019060 PMCID: PMC4073229 DOI: 10.3389/fonc.2014.00166] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/10/2014] [Indexed: 11/23/2022] Open
Abstract
Purpose: Pheochromocytomas (PCCs) are neuroendocrine tumors arising from the adrenal medulla or as paraganglioma (PGL) from extra-adrenal sites. While usually benign, a small fraction is malignant. Multi-modality therapy is used in treating malignant disease; however, little data exist on the role of external beam radiation therapy (EBRT). In this retrospective review, we assessed response to EBRT in malignant PCCs or PGLs. Methods and Materials: Records of patients treated at the National Institutes of Health who received EBRT between 1990 and 2012 were studied. Patients were assessed for symptomatic control, biochemical response, local and distant control by response evaluation criteria in solid tumors v1.1 or stable disease on imaging reports, toxicity by radiation therapy oncology group (RTOG) criteria, and survival. Results: There were 24 patients treated who received EBRT to lesions of the abdomen (n = 3), central nervous system (n = 4), and bone (n = 40). Lesions were treated with 3D conformal EBRT to a mean dose of 31.8 Gy in 3.3 Gy fractions, or fractionated stereotactic radiosurgery to 21.9 Gy in 13.6 Gy fractions. Patients experienced acute (n = 15) and late (n = 2) RTOG toxicities; no patient experienced acute toxicity ≥4 or late toxicity ≥2. Symptomatic control was achieved in 81.1% of lesions. Stable radiographic response was achieved in 86.7% of lesions with progression in 13%. Distant progression was observed overall in 75% of patients and average survival was 52.4 months. Conclusion: Malignant PCC and PGL often do not respond well to current systemic therapies. In these cases, EBRT can be considered in patients with symptomatic, localized disease progression.
Collapse
Affiliation(s)
- Jennifer Vogel
- NIH, Radiation Oncology Branch, National Cancer Institute , Bethesda, MD , USA
| | - Aileen Sia Atanacio
- NIH, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institutes of Child Health and Human Development , Bethesda, MD , USA
| | - Tamara Prodanov
- NIH, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institutes of Child Health and Human Development , Bethesda, MD , USA
| | | | - Karen Adams
- NIH, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institutes of Child Health and Human Development , Bethesda, MD , USA
| | - Victoria Martucci
- NIH, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institutes of Child Health and Human Development , Bethesda, MD , USA
| | - Kevin Camphausen
- NIH, Radiation Oncology Branch, National Cancer Institute , Bethesda, MD , USA
| | - Antonio Tito Fojo
- NIH, Medical Oncology Branch, National Cancer Institute , Bethesda, MD , USA
| | - Karel Pacak
- NIH, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institutes of Child Health and Human Development , Bethesda, MD , USA
| | - Aradhana Kaushal
- NIH, Radiation Oncology Branch, National Cancer Institute , Bethesda, MD , USA
| |
Collapse
|
46
|
van Hulsteijn LT, Niemeijer ND, Dekkers OM, Corssmit EPM. (131)I-MIBG therapy for malignant paraganglioma and phaeochromocytoma: systematic review and meta-analysis. Clin Endocrinol (Oxf) 2014; 80:487-501. [PMID: 24118038 DOI: 10.1111/cen.12341] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/14/2013] [Accepted: 02/23/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND (131)I-MIBG therapy can be used for palliative treatment of malignant paraganglioma and phaeochromocytoma. The main objective of this study was to perform a systematic review and meta-analysis assessing the effect of (131)I-MIBG therapy on tumour volume in patients with malignant paraganglioma/phaeochromocytoma. METHODS A literature search was performed in December 2012 to identify potentially relevant studies. Main outcomes were the pooled proportions of complete response, partial response and stable disease after radionuclide therapy. A meta-analysis was performed with an exact likelihood approach using a logistic regression with a random effect at the study level. Pooled proportions with 95% confidence intervals (CI) were reported. RESULTS Seventeen studies concerning a total of 243 patients with malignant paraganglioma/phaeochromocytoma were treated with (131)I-MIBG therapy. The mean follow-up ranged from 24 to 62 months. A meta-analysis of the effect of (131)I-MIBG therapy on tumour volume showed pooled proportions of complete response, partial response and stable disease of, respectively, 0·03 (95% CI: 0·06-0·15), 0·27 (95% CI: 0·19-0·37) and 0·52 (95% CI: 0·41-0·62) and for hormonal response 0·11 (95% CI: 0·05-0·22), 0·40 (95% CI: 0·28-0·53) and 0·21 (95% CI: 0·10-0·40), respectively. Separate analyses resulted in better results in hormonal response for patients with paraganglioma than for patients with phaeochromocytoma. CONCLUSIONS Data on the effects of (131)I-MIBG therapy on malignant paraganglioma/phaeochromocytoma suggest that stable disease concerning tumour volume and a partial hormonal response can be achieved in over 50% and 40% of patients, respectively, treated with (131)I-MIBG therapy. It cannot be ruled out that stable disease reflects not only the effect of MIBG therapy, but also (partly) the natural course of the disease.
Collapse
Affiliation(s)
- L T van Hulsteijn
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
47
|
Hodin R, Lubitz C, Phitayakorn R, Stephen A. Diagnosis and management of pheochromocytoma. Curr Probl Surg 2014; 51:151-87. [DOI: 10.1067/j.cpsurg.2013.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/27/2013] [Indexed: 12/21/2022]
|
48
|
Choi JDW, Tucker KM, Lee TT, Chong GC. Hereditary paraganglioma-pheochromocytoma syndromes associated with SDHD and RET mutations. Head Neck 2014; 36:E99-E102. [PMID: 24375508 DOI: 10.1002/hed.23598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/13/2013] [Accepted: 12/20/2013] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Hereditary paraganglioma-pheochromocytoma syndromes (PGL/PCC) are rare tumors arising from neuroendocrine cells. METHODS AND RESULTS The proband, a 59-year-old white man and his 42-year-old elder son had a medical history of bilateral carotid body PGL and both presented for treatment of abdominal PGLs. His 36-year-old daughter had excision of recurrent malignant carotid body PGL and vertebral metastasis. His 33-year-old youngest son presented for excision of a unilateral carotid body PGL. All 4 members had succinate dehydrogenase subunit D (SDHD) mutations, whereas the proband and youngest son also had concurrent rearranged during transfection (RET) mutation. CONCLUSION This is the first report of PGL/PCC with SDHD and RET mutations. The role of the RET gene as a modifier remains speculative. Additionally, the family pedigree suggests maternal inheritance of disease from the probands' paternal grandmother. Clinicians should refer PGL/PCC families for mutation analysis as well as being alert to changes in the classification of mutations.
Collapse
Affiliation(s)
- Joseph Do Woong Choi
- Academic Unit of Surgery, Australian National University Medical School, The Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | | | | | | |
Collapse
|
49
|
Martucci VL, Pacak K. Pheochromocytoma and paraganglioma: diagnosis, genetics, management, and treatment. Curr Probl Cancer 2014; 38:7-41. [PMID: 24636754 DOI: 10.1016/j.currproblcancer.2014.01.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
50
|
Lowery AJ, Walsh S, McDermott EW, Prichard RS. Molecular and therapeutic advances in the diagnosis and management of malignant pheochromocytomas and paragangliomas. Oncologist 2013; 18:391-407. [PMID: 23576482 DOI: 10.1634/theoncologist.2012-0410] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare catecholamine-secreting tumors derived from chromaffin cells originating in the neural crest. These tumors represent a significant diagnostic and therapeutic challenge because the diagnosis of malignancy is frequently made in retrospect by the development of metastatic or recurrent disease. Complete surgical resection offers the only potential for cure; however, recurrence can occur even after apparently successful resection of the primary tumor. The prognosis for malignant disease is poor because traditional treatment modalities have been limited. The last decade has witnessed exciting discoveries in the study of PCCs and PGLs; advances in molecular genetics have uncovered hereditary and germline mutations of at least 10 genes that contribute to the development of these tumors, and increasing knowledge of genotype-phenotype interactions has facilitated more accurate determination of malignant potential. Elucidating the molecular mechanisms responsible for malignant transformation in these tumors has opened avenues of investigation into targeted therapeutics that show promising results. There have also been significant advances in functional and radiological imaging and in the surgical approach to adrenalectomy, which remains the mainstay of treatment for PCC. In this review, we discuss the currently available diagnostic and therapeutic options for patients with malignant PCCs and PGLs and detail the molecular rationale and clinical evidence for novel and emerging diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Aoife J Lowery
- Department of Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|