1
|
Savvidis C, Kallistrou E, Kouroglou E, Dionysopoulou S, Gavriiloglou G, Ragia D, Tsiama V, Proikaki S, Belis K, Ilias I. Circadian rhythm disruption and endocrine-related tumors. World J Clin Oncol 2024; 15:818-834. [PMID: 39071458 PMCID: PMC11271730 DOI: 10.5306/wjco.v15.i7.818] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This review delved into the intricate relationship between circadian clocks and physiological processes, emphasizing their critical role in maintaining homeostasis. Orchestrated by interlocked clock genes, the circadian timekeeping system regulates fundamental processes like the sleep-wake cycle, energy metabolism, immune function, and cell proliferation. The central oscillator in the hypothalamic suprachiasmatic nucleus synchronizes with light-dark cycles, while peripheral tissue clocks are influenced by cues such as feeding times. Circadian disruption, linked to modern lifestyle factors like night shift work, correlates with adverse health outcomes, including metabolic syndrome, cardiovascular diseases, infections, and cancer. We explored the molecular mechanisms of circadian clock genes and their impact on metabolic disorders and cancer pathogenesis. Specific associations between circadian disruption and endocrine tumors, spanning breast, ovarian, testicular, prostate, thyroid, pituitary, and adrenal gland cancers, are highlighted. Shift work is associated with increased breast cancer risk, with PER genes influencing tumor progression and drug resistance. CLOCK gene expression correlates with cisplatin resistance in ovarian cancer, while factors like aging and intermittent fasting affect prostate cancer. Our review underscored the intricate interplay between circadian rhythms and cancer, involving the regulation of the cell cycle, DNA repair, metabolism, immune function, and the tumor microenvironment. We advocated for integrating biological timing into clinical considerations for personalized healthcare, proposing that understanding these connections could lead to novel therapeutic approaches. Evidence supports circadian rhythm-focused therapies, particularly chronotherapy, for treating endocrine tumors. Our review called for further research to uncover detailed connections between circadian clocks and cancer, providing essential insights for targeted treatments. We emphasized the importance of public health interventions to mitigate lifestyle-related circadian disruptions and underscored the critical role of circadian rhythms in disease mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Christos Savvidis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Efthymia Kallistrou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Eleni Kouroglou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Sofia Dionysopoulou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | | | - Dimitra Ragia
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Vasiliki Tsiama
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Stella Proikaki
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Konstantinos Belis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Ioannis Ilias
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| |
Collapse
|
2
|
Milinkovic M, Soldatovic I, Zivaljevic V, Bozic V, Zivotic M, Tatic S, Dundjerovic D. Comprehensive Investigation of Angiogenesis, PASS Score and Immunohistochemical Factors in Risk Assessment of Malignancy for Paraganglioma and Pheochromocytoma. Diagnostics (Basel) 2024; 14:849. [PMID: 38667494 PMCID: PMC11049119 DOI: 10.3390/diagnostics14080849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
A challenging task in routine practice is finding the distinction between benign and malignant paragangliomas and pheochromocytomas. The aim of this study is to conduct a comparative analysis of angiogenesis by assessing intratumoral microvascular density (MVD) with immunohistochemical (IHC) markers (CD31, CD34, CD105, ERG), and S100 immunoreactivity, Ki67 proliferative index, succinate dehydrogenase B (SDHB) expressiveness, tumor size with one the most utilized score Pheochromocytoma of Adrenal Gland Scales Score (PASS), using tissue microarray (TMA) with 115 tumor samples, 61 benign (PASS < 4) and 54 potentially malignant (PASS ≥ 4). We found no notable difference between intratumoral MVD and potentially malignant behavior. The group of potentially malignant tumors is significantly larger in size, has lower intratumoral MVD, and a decreased number of S100 labeled sustentacular cells. Both groups have low proliferative activity (mean Ki67 is 1.02 and 1.22, respectively). Most tumors maintain SDHB expression, only 6 cases (5.2%) showed a loss of expression (4 of them in PASS < 4 group and 2 in PASS ≥ 4). PASS score is easily available for assessment and complemented with markers of biological behavior to complete the risk stratification algorithm. Size is directly related to PASS score and malignancy. Intratumoral MVD is extensively developed but it is not crucial in evaluating the malignant potential.
Collapse
Affiliation(s)
- Marija Milinkovic
- Department of Pathology, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Ivan Soldatovic
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vladan Zivaljevic
- Clinic for Endocrine Surgery, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Vesna Bozic
- Department of Pathology, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Maja Zivotic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.); (S.T.); (D.D.)
| | - Svetislav Tatic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.); (S.T.); (D.D.)
| | - Dusko Dundjerovic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.Z.); (S.T.); (D.D.)
| |
Collapse
|
3
|
Mancini M, Buffet A, Porte B, Amar L, Lussey-Lepoutre C, Crinière L, Baudin E, Meatchi T, Gimenez-Roqueplo AP, Favier J, Burnichon N. EPAS1-mutated paragangliomas associated with haemoglobin disorders. Br J Haematol 2024; 204:1054-1060. [PMID: 38195958 DOI: 10.1111/bjh.19278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024]
Abstract
We report a large series of 40 patients presenting EPAS1-mutated paraganglioma (PGL) in whom we investigated a cause underlying chronic hypoxia. Four patients suffered from hypoxaemic heart disease. In patients with available haemoglobin electrophoresis results, 59% presented with a haemoglobin disorder, including six with sickle cell disease, five with sickle cell trait and two with heterozygous haemoglobin C disease. Histological and transcriptomic characterization of EPAS1 tumours revealed increased angiogenesis and high similarities with pseudohypoxic PGLs caused by VHL gene mutations. Sickle haemoglobinopathy carriers could thus be at increased risk for developing EPAS1-PGLs, which should be taken into account in their management and surveillance.
Collapse
Affiliation(s)
- Maxence Mancini
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Alexandre Buffet
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Baptiste Porte
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Laurence Amar
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Service d'Hypertension artérielle, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Charlotte Lussey-Lepoutre
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Service de Médecine Nucléaire, Sorbonne Université, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Lise Crinière
- Service d'endocrinologie, CHRU Bretonneau, Tours, France
| | - Eric Baudin
- Service de Médecine Nucléaire, Gustave Roussy, Villejuif, France
| | - Tchao Meatchi
- Service d'anatomie pathologique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| | - Judith Favier
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Nelly Burnichon
- Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
- Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
4
|
Smith J, Barnett E, Rodger EJ, Chatterjee A, Subramaniam RM. Neuroendocrine Neoplasms: Genetics and Epigenetics. PET Clin 2023; 18:169-187. [PMID: 36858744 DOI: 10.1016/j.cpet.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Neuroendocrine neoplasms (NENs) are a group of rare, heterogeneous tumors of neuroendocrine cell origin, affecting a range of different organs. The clinical management of NENs poses significant challenges, as tumors are often diagnosed at an advanced stage where overall survival remains poor with current treatment regimens. In addition, a host of complex and often unique molecular changes underpin the pathobiology of each NEN subtype. Exploitation of the unique genetic and epigenetic signatures driving each NEN subtype provides an opportunity to enhance the diagnosis, treatment, and monitoring of NEN in an emerging era of individualized medicine.
Collapse
Affiliation(s)
- Jim Smith
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Te Whatu Ora - Southern, Dunedin Public Hospital, 270 Great King Street, PO Box 913, Dunedin, New Zealand.
| | - Edward Barnett
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Euan J Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Rathan M Subramaniam
- Department of Medicine, Otago Medical School, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Radiology, Duke University, 2301 Erwin Rd, BOX 3808, Durham, NC 27705, USA
| |
Collapse
|
5
|
Noureldine MHA, Shimony N, Jallo GI. Benign Spinal Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:583-606. [PMID: 37452955 DOI: 10.1007/978-3-031-23705-8_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Benign spinal intradural tumors are relatively rare and include intramedullary tumors with a favorable histology such as low-grade astrocytomas and ependymomas, as well as intradural extramedullary tumors such as meningiomas and schwannomas. The effect on the neural tissue is usually a combination of mass effect and neuronal involvement in cases of infiltrative tumors. The new understanding of molecular profiling of different tumors allowed us to better define central nervous system tumors and tailor treatment accordingly. The mainstay of management of many intradural spinal tumors is maximal safe surgical resection. This goal is more achievable with intradural extramedullary tumors; yet, with a meticulous surgical approach, many of the intramedullary tumors are amenable for safe gross-total or near-total resection. The nature of these tumors is benign; hence, a different way to measure outcome success is pursued and usually depends on functional rather than oncological or survival outcomes.
Collapse
Affiliation(s)
- Mohammad Hassan A Noureldine
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Institute for Brain Protection Sciences, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
| | - Nir Shimony
- Institute of Neuroscience, Geisinger Medical Center, Geisinger Commonwealth School of Medicine, Danville, PA, USA
- Institute for Brain Protections Sciences, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
- Department of Surgery, St Jude Children's Research Hospital, Memphis, USA
| | - George I Jallo
- Institute for Brain Protections Sciences, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA.
| |
Collapse
|
6
|
|
7
|
Sousa D, Pereira SS, Pignatelli D. Modulation of Autophagy in Adrenal Tumors. Front Endocrinol (Lausanne) 2022; 13:937367. [PMID: 35966083 PMCID: PMC9373848 DOI: 10.3389/fendo.2022.937367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/15/2022] [Indexed: 01/18/2023] Open
Abstract
Adrenal masses are one of the most common tumors in humans. The majority are benign and non-functioning and therefore do not require immediate treatment. In contrast, the rare adrenal malignant tumors are often highly aggressive and with poor prognosis. Besides usually being detected in advanced stages, often already with metastases, one of the reasons of the unfavorable outcome of the patients with adrenal cancer is the absence of effective treatments. Autophagy is one of the intracellular pathways targeted by several classes of chemotherapeutics. Mitotane, the most commonly used drug for the treatment of adrenocortical carcinoma, was recently shown to also modulate autophagy. Autophagy is a continuous programmed cellular process which culminates with the degradation of cellular organelles and proteins. However, being a dynamic mechanism, understanding the autophagic flux can be highly complex. The role of autophagy in cancer has been described paradoxically: initially described as a tumor pro-survival mechanism, different studies have been showing that it may result in other outcomes, namely in tumor cell death. In adrenal tumors, this dual role of autophagy has also been addressed in recent years. Studies reported both induction and inhibition of autophagy as a treatment strategy of adrenal malignancies. Importantly, most of these studies were performed using cell lines. Consequently clinical studies are still required. In this review, we describe what is known about the role of autophagy modulation in treatment of adrenal tumors. We will also highlight the aspects that need further evaluation to understand the paradoxical role of autophagy in adrenal tumors.
Collapse
Affiliation(s)
- Diana Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Sofia S. Pereira
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Duarte Pignatelli
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Endocrinology, Centro Hospitalar e Universitário de S. João, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine of the University of Porto, Porto, Portugal
| |
Collapse
|
8
|
Yoda RA, Cimino PJ. Neuropathologic features of central nervous system hemangioblastoma. J Pathol Transl Med 2022; 56:115-125. [PMID: 35501672 PMCID: PMC9119802 DOI: 10.4132/jptm.2022.04.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/13/2022] [Indexed: 12/04/2022] Open
Abstract
Hemangioblastoma is a benign, highly vascularized neoplasm of the central nervous system (CNS). This tumor is associated with loss of function of the VHL gene and demonstrates frequent occurrence in von Hippel-Lindau (VHL) disease. While this entity is designated CNS World Health Organization grade 1, due to its predilection for the cerebellum, brainstem, and spinal cord, it is still an important cause of morbidity and mortality in affected patients. Recognition and accurate diagnosis of hemangioblastoma is essential for the practice of surgical neuropathology. Other CNS neoplasms, including several tumors associated with VHL disease, may present as histologic mimics, making diagnosis challenging. We outline key clinical and radiologic features, pathophysiology, treatment modalities, and prognostic information for hemangioblastoma, and provide a thorough review of the gross, microscopic, immunophenotypic, and molecular features used to guide diagnosis.
Collapse
Affiliation(s)
- Rebecca A. Yoda
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, Division of Cytopathology, University of Washington, Seattle, WA, USA
- Corresponding Author: Rebecca A. Yoda, MD, Department of Laboratory Medicine and Pathology, University of Washington, 325 9th Avenue, Box 359791, Seattle, WA 98104-2499, USA Tel: +1-206-744-3145, Fax: +1-206-744-8240, E-mail:
| | - Patrick J. Cimino
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
9
|
Hassan Nelson L, Fuentes-Bayne H, Yin J, Asmus E, Ryder M, Morris JC, Hilger CR, Bible KC, Chintakuntlawar AV, Rao SN. Lenvatinib as a Therapeutic Option in Unresectable Metastatic Pheochromocytoma and Paragangliomas. J Endocr Soc 2022; 6:bvac044. [PMID: 35402763 PMCID: PMC8989149 DOI: 10.1210/jendso/bvac044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Context
Metastatic pheochromocytomas and paragangliomas (mPPGL) are rare vascular neuroendocrine tumors that highly express vascular growth factors. Systemic treatment options in cases of unresectable multi-site disease are limited. Multikinase inhibitors, that inhibit angiogenesis, such as lenvatinib, have proven effective in several other malignancies, and may be a viable option for mPPGL.
Objective
To evaluate the efficacy of lenvatinib as salvage therapy in mPPGLs.
Design
Retrospective analysis of mPPGL patients who received lenvatinib from 2015-2020.
Setting
Tertiary referral center
Patients
Patients ≥ 18 years with mPPGL who were treated with lenvatinib were included.
Intervention
Patients were started on lenvatinib 20 mg daily and dose was adjusted according to tolerance or disease progression.
Results
11 patients were included. Median treatment duration was 14.7 months (95% CI: 2.3-NE). Treatment was discontinued due to disease progression, adverse events, or death. Overall survival at 12 months was 80.8% (95% CI: 42.3-94.9%) but its median was not reached. Median progression free survival was 14.7 months (CI 95%: 1.7-NE). Among the 8 patients with measurable disease, overall response rate was 63% as 5/8 experienced a partial response and 3/8 had stable disease. Worsening hypertension and anemia were the most common adverse events (AE).
Conclusions
Lenvatinib may be a viable treatment option for mPPGL, though at the potential risk of worsening hypertension. Larger, multi-center studies are needed to better characterize treatment efficacy.
Collapse
Affiliation(s)
| | | | - Jun Yin
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Erik Asmus
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Mabel Ryder
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | - John C Morris
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | - Sarika N Rao
- Division of Endocrinology, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
10
|
Spada A, Mantovani G, Lania AG, Treppiedi D, Mangili F, Catalano R, Carosi G, Sala E, Peverelli E. Pituitary Tumors: Genetic and Molecular Factors Underlying Pathogenesis and Clinical Behavior. Neuroendocrinology 2022; 112:15-33. [PMID: 33524974 DOI: 10.1159/000514862] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022]
Abstract
Pituitary neuroendocrine tumors (PitNETs) are the most common intracranial neoplasms. Although generally benign, they can show a clinically aggressive course, with local invasion, recurrences, and resistance to medical treatment. No universally accepted biomarkers of aggressiveness are available yet, and predicting clinical behavior of PitNETs remains a challenge. In rare cases, the presence of germline mutations in specific genes predisposes to PitNET formation, as part of syndromic diseases or familial isolated pituitary adenomas, and associates to more aggressive, invasive, and drug-resistant tumors. The vast majority of cases is represented by sporadic PitNETs. Somatic mutations in the α subunit of the stimulatory G protein gene (gsp) and in the ubiquitin-specific protease 8 (USP8) gene have been recognized as pathogenetic factors in sporadic GH- and ACTH-secreting PitNETs, respectively, without an association with a worse clinical phenotype. Other molecular factors have been found to significantly affect PitNET drug responsiveness and invasive behavior. These molecules are cytoskeleton and/or scaffold proteins whose alterations prevent proper functioning of the somatostatin and dopamine receptors, targets of medical therapy, or promote the ability of tumor cells to invade surrounding tissues. The aim of the present review is to provide an overview of the genetic and molecular alterations that can contribute to determine PitNET clinical behavior. Understanding subcellular mechanisms underlying pituitary tumorigenesis and PitNET clinical phenotype will hopefully lead to identification of new potential therapeutic targets and new markers predicting the behavior and the response to therapeutic treatments of PitNETs.
Collapse
Affiliation(s)
- Anna Spada
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea G Lania
- Endocrinology, Diabetology and Medical Andrology Unit, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Donatella Treppiedi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Mangili
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Rosa Catalano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giulia Carosi
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Sala
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy,
| |
Collapse
|
11
|
Doyle C, Bolger J, Conneely JB, Walsh KP. Paraganglioma in an adolescent awaiting a cardiac transplant due to unrepaired congenital cyanotic heart disease and a univentricular heart: the first reported case. BMJ Case Rep 2021; 14:14/5/e241804. [PMID: 34035024 DOI: 10.1136/bcr-2021-241804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We report a case of a 16-year-old adolescent male born with univentricular congenital cyanotic heart disease (CCHD) who was diagnosed with an incidental paraganglioma while awaiting a cardiac transplant. The coexistence of paraganglioma and univentricular CCHD is very rare, with no previous cases described in the literature of a patient concurrently requiring a cardiac transplant. The complex physiology associated with a common atrium, common ventricle, aortopulmonary lung perfusion and a hypoplastic left lung rendered our patient extremely vulnerable to catecholamine-mediated effects of preload, contractility and afterload. The interactions and interdependence between these systems provided unique difficulties for perioperative management with serious implications for prospective cardiac transplant.
Collapse
Affiliation(s)
- Conor Doyle
- Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jarlath Bolger
- General Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - John B Conneely
- Department of Hepatobiliary Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Kevin P Walsh
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Xing J, Cheng Y, Ying H, Guan M, Jia N, Bai C. Systemic treatment of a metastatic carotid body tumor: A case report and literature review. Medicine (Baltimore) 2020; 99:e22811. [PMID: 33217796 PMCID: PMC7676560 DOI: 10.1097/md.0000000000022811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/08/2020] [Accepted: 09/21/2020] [Indexed: 11/30/2022] Open
Abstract
RATIONALE Carotid body tumors (CBTs) are head and neck paragangliomas (PGLs) with a low incidence of distant metastasis. To date, only a few metastatic cases treated with detailed systemic therapy are reported and effective management is still inconclusive. Herein, we reported a metastatic CBT case with systemic therapy and reviewed the reported systemic treatment. PATIENT CONCERNS A 56-year-old man noticed multiple painless nodules on the right side of the neck and developed debilitating chest and back pain 7 years after the CBT resection. DIAGNOSES Widespread bone and lymph nodes CBT metastases. INTERVENTIONS Biopsies of the enlarged lymph nodes confirmed the diagnosis of metastatic CBT and 18F-FDG PET-CT detected multiple right cervical lymph nodes and bone metastases. 24 cycles of cyclophosphamide, vincristine and dacarbazine (CVD) chemotherapy were given since May 2016 to Jul 2018 and dacarbazine maintenance therapy was given in the next 15 months follow-up period. OUTCOMES Partial remission was achieved according to the Response Evaluation in Criteria in Solid Tumors 1.1 criteria. A prominent control in the metastatic lesions were also observed in 18F-FDG PET-CT scan. LESSONS Evidence for systemic management of metastatic CBTs is mainly based on studies of PGLs and pheochromocytoma. According to our review on metastatic CBT cases treated with systemic therapy from 1981 to 2018, chemotherapy, especially the CVD regimen, was a common reported management. In SDHB mutated patients, sunitinib and temozolomide could also be considered.
Collapse
|
13
|
Cummings JJ, Siegrist KK, Deegan RJ, Solórzano CC, Eagle SS. Robotic Adrenalectomy for Pheochromocytoma in a Patient with Fontan Physiology. J Cardiothorac Vasc Anesth 2020; 34:2446-2451. [PMID: 32434722 DOI: 10.1053/j.jvca.2020.02.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Jared J Cummings
- Division of Cardiothoracic Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Kara K Siegrist
- Division of Cardiothoracic Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Robert J Deegan
- Division of Cardiothoracic Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Carmen C Solórzano
- Division of Cardiothoracic Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan S Eagle
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
14
|
Liu Y, Pang Y, Zhu B, Uher O, Caisova V, Huynh TT, Taieb D, Hadrava Vanova K, Ghayee HK, Neuzil J, Levine M, Yang C, Pacak K. Therapeutic Targeting of SDHB-Mutated Pheochromocytoma/Paraganglioma with Pharmacologic Ascorbic Acid. Clin Cancer Res 2020; 26:3868-3880. [PMID: 32152203 DOI: 10.1158/1078-0432.ccr-19-2335] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/09/2019] [Accepted: 03/04/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Pheochromocytomas and paragangliomas (PCPG) are usually benign neuroendocrine tumors. However, PCPGs with mutations in the succinate dehydrogenase B subunit (SDHB) have a poor prognosis and frequently develop metastatic lesions. SDHB-mutated PCPGs exhibit dysregulation in oxygen metabolic pathways, including pseudohypoxia and formation of reactive oxygen species, suggesting that targeting the redox balance pathway could be a potential therapeutic approach. EXPERIMENTAL DESIGN We studied the genetic alterations of cluster I PCPGs compared with cluster II PCPGs, which usually present as benign tumors. By targeting the signature molecular pathway, we investigated the therapeutic effect of ascorbic acid on PCPGs using in vitro and in vivo models. RESULTS By investigating PCPG cells with low SDHB levels, we show that pseudohypoxia resulted in elevated expression of iron transport proteins, including transferrin (TF), transferrin receptor 2 (TFR2), and the divalent metal transporter 1 (SLC11A2; DMT1), leading to iron accumulation. This iron overload contributed to elevated oxidative stress. Ascorbic acid at pharmacologic concentrations disrupted redox homeostasis, inducing DNA oxidative damage and cell apoptosis in PCPG cells with low SDHB levels. Moreover, through a preclinical animal model with PCPG allografts, we demonstrated that pharmacologic ascorbic acid suppressed SDHB-low metastatic lesions and prolonged overall survival. CONCLUSIONS The data here demonstrate that targeting redox homeostasis as a cancer vulnerability with pharmacologic ascorbic acid is a promising therapeutic strategy for SDHB-mutated PCPGs.
Collapse
Affiliation(s)
- Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Ying Pang
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Boqun Zhu
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.,Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.,Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Thanh-Truc Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Katerina Hadrava Vanova
- Molecular Therapy Group, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Hans Kumar Ghayee
- Department of Internal Medicine, Division of Endocrinology, University of Florida College of Medicine and Malcom Randall VA Medical Center, Gainesville, Florida
| | - Jiri Neuzil
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic.,Mitochondria, Apoptosis and Cancer Research Group, School of Medical Science and Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.
| |
Collapse
|
15
|
Ng P, Deepak D, Teo L, Low TT. Asymptomatic phaeochromocytoma in a patient with Holt-Oram syndrome: a case report. EUROPEAN HEART JOURNAL-CASE REPORTS 2020; 3:1-5. [PMID: 32099963 PMCID: PMC7026609 DOI: 10.1093/ehjcr/ytz206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/29/2019] [Accepted: 10/21/2019] [Indexed: 11/19/2022]
Abstract
Background Holt-Oram syndrome (HOS) is a rare congenital disease that affects the heart and upper limbs. Phaeochromocytoma, a catecholamine-secreting tumour, is a rare neuroendocrine disorder. We present an interesting case presentation of these two rare disorders in a patient who was asymptomatic for phaeochromocytoma. Case summary A 28-year-old woman who was diagnosed at birth with HOS, presented to the hospital with heart failure. She has a past medical history of corrected cyanotic congenital heart disease. She presented with dyspnoea but she did not have headaches, tremors, or diaphoresis. Cardiac magnetic resonance scan was done to investigate the cause of her heart failure and revealed right ventricular systolic dysfunction and a suspicious adrenal lesion. Magnetic resonance imaging adrenal confirmed the presence of the adrenal lesion and concerns were raised for a possible phaeochromocytoma. Biochemical tests showed raised plasma free metanephrine levels. Gallium-68 DOTA positron emission tomography scan showed intense right adrenal gland uptake in keeping with diagnosis of phaeochromocytoma. Discussion Phaeochromocytoma appears to be more prevalent in patients who are in a chronic hypoxic state. This hypoxic state has been postulated to cause the proliferation of adrenal tissue and therefore the formation of phaeochromocytomas. The hypoxia-inducing factor, which is increased in patients with phaeochromocytoma, has been identified as one of the key factors driving this process as it modulates genes that regulate angiogenesis and proliferation. Congenital heart defects seen in HOS can progress to cyanotic heart disease if left uncorrected and may have been the driver for the development of phaeochromocytoma in our patient.
Collapse
Affiliation(s)
- Perryn Ng
- National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Doddabele Deepak
- National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Lynette Teo
- National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Ting Ting Low
- National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
16
|
Gao X, Yamazaki Y, Pecori A, Tezuka Y, Ono Y, Omata K, Morimoto R, Nakamura Y, Satoh F, Sasano H. Histopathological Analysis of Tumor Microenvironment and Angiogenesis in Pheochromocytoma. Front Endocrinol (Lausanne) 2020; 11:587779. [PMID: 33244312 PMCID: PMC7685215 DOI: 10.3389/fendo.2020.587779] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Pheochromocytomas (PHEOs) are relatively rare catecholamine-producing tumors derived from adrenal medulla. Tumor microenvironment (TME) including neoangiogenesis has been explored in many human neoplasms but not necessarily in PHEOs. Therefore, in this study, we examined tumor infiltrating lymphocytes (CD4 and CD8), tumor associated macrophages (CD68 and CD163), sustentacular cells (S100p), and angiogenic markers (CD31 and areas of intratumoral hemorrhage) in 39 cases of PHEOs in the quantitative fashion. We then compared the results with pheochromocytoma of the adrenal gland scaled score (PASS), grading system for pheochromocytoma and paraganglioma (GAPP) and the status of intra-tumoral catecholamine-synthesizing enzymes (TH, DDC, and PNMT) as well as their clinicopathological factors. Intratumoral CD8 (p = 0.0256), CD31 (p = 0.0400), and PNMT (p = 0.0498) status was significantly higher in PHEOs with PASS <4 than PASS ≧4. In addition, intratumoral CD8+ lymphocytes were also significantly more abundant in well-than moderately differentiated PHEO according to GAPP score (p = 0.0108) and inversely correlated with tumor size (p = 0.0257). Intratumoral CD68+ cells were significantly higher in PHEOs with regular or normal histological patterns than those not (p = 0.0370) and inversely correlated with tumor size (p = 0.0457). The status of CD163 was significantly positively correlated with that of CD8 positive cells (p = 0.0032). The proportion of intratumoral hemorrhage areas was significantly higher in PHEOs with PASS ≧4 (p = 0.0172). DDC immunoreactivity in tumor cells was significantly positively correlated with PASS score (p = 0.0356) and TH status was significantly higher in PHEOs harboring normal histological patterns (p = 0.0236) and cellular monotony (p = 0.0219) than those not. Results of our present study did demonstrate that abundant CD8+ and CD68+ cells could represent a histologically low-scored tumor. In particular, PHEOs with increased intratumoral hemorrhage should be considered rather malignant. In addition, abnormal catecholamine-producing status of tumor cells such as deficient PNMT and TH and increased DDC could also represent more aggressive PHEOs.
Collapse
Affiliation(s)
- Xin Gao
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alessio Pecori
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuta Tezuka
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yoshikiyo Ono
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Kei Omata
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Fumitoshi Satoh
- Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Hironobu Sasano,
| |
Collapse
|
17
|
Agarwal S, Jindal I, Balazs A, Paul D. Catecholamine-Secreting Tumors in Pediatric Patients With Cyanotic Congenital Heart Disease. J Endocr Soc 2019; 3:2135-2150. [PMID: 31687640 PMCID: PMC6821216 DOI: 10.1210/js.2019-00226] [Citation(s) in RCA: 2] [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: 06/14/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Catecholamine-secreting tumors are rare among the pediatric population but are increasingly being reported in children with sustained hypoxia secondary to cyanotic congenital heart disease (CCHD). With this review, we report the clinical characteristics of these tumors in children with CCHD. The articles included in the present review were identified using PubMed through February 2019. A manual search of the references retrieved from relevant articles was also performed. Pheochromocytomas and paragangliomas (PPGL) in children are commonly associated with high-risk germline or somatic mutations. There is evidently a higher risk of tumorigenesis in children with CCHD as compared with the general pediatric population, even in the absence of susceptible gene mutations. This is due to molecular mechanisms involving the aberrant activation of hypoxia-response elements, likely secondary to sustained hypoxemia, resulting in tumorigenesis. Due to overlapping symptoms with CCHD, the diagnosis of PPGL may be delayed or missed in these patients. We studied all previously reported PPGL cases in children with CCHD and reviewed phenotypic and biochemical features to assess for contributing factors in tumorigenesis. Larger studies are needed to help determine other potential predisposing factors and to establish screening guidelines in this high-risk population. A delay in diagnosis of the PPGL tumors can lead to exacerbation of cardiac failure, and therefore early diagnosis and intervention may provide better outcomes in these patients, necessitating the need for regular surveillance. We recommend routine biochemical screening in patients with sustained hypoxia secondary to CCHD.
Collapse
Affiliation(s)
- Swashti Agarwal
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Ishita Jindal
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Andrea Balazs
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - David Paul
- Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
18
|
Genetic and epigenetic differences of benign and malignant pheochromocytomas and paragangliomas (PPGLs). Endocr Regul 2019; 52:41-54. [PMID: 29453919 DOI: 10.2478/enr-2018-0006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are tumors arising from the adrenal medulla and sympathetic/parasympathetic paraganglia, respectively. According to Th e Cancer Genome Atlas (TCGA), approximately 40% of PPGLs are due to germ line mutations in one of 16 susceptibility genes, and a further 30% are due to somatic alterations in at least seven main genes (VHL, EPAS1, CSDE1, MAX, HRAS, NF1, RET, and possibly KIF1B). Th e diagnosis of malignant PPGL was straight forward in most cases as it was defined as presence of PPGL in non-chromaffin tissues. Accordingly, there is an extreme need for new diagnostic marker(s) to identify tumors with malignant prospective. Th e aim of this study was to review all suggested genetic and epigenetic alterations that are remarkably different between benign and malignant PPGLs. It seems that more than two genetic mutation clusters in PPGLs and other genetic and methylation biomarkers could be targeted for malignancy discrimination in different studies.
Collapse
|
19
|
Abstract
Pheochromocytoma/paraganglioma is an exceedingly rare tumour, thought to share an association with cyanotic CHD. This association is thought to be a result of chronic hypoxaemia (Antonio et al, Revista Española de Cardiología (English Edition) 2017; 70: 673-675; Folger et al, Circulation 1964; 29: 750-757; Opotowsky et al, J Clin Endocrinol Metab 2015; 100: 1325-1334) We report two cases of paraganglioma over a 4-year period in patients with hypoplastic left heart syndrome who had undergone Fontan completion by ages 2 and 4. Based on a very small number of reported cases of CHD, the mechanism of tumourigenesis is unclear. It is imperative that cases associated with CHD continue to be reported so that we may learn more about the pathogenesis and epidemiology of this entity.
Collapse
|
20
|
Goroshi M, Jadhav SS, Sarathi V, Lila AR, Patil VA, Shah R, Hira P, Sharma R, Goroshi S, Fernandes G, Rojekar A, Dalvi A, Bakshi G, Prakash G, Shah NS, Bandgar TR. Radiological differentiation of phaeochromocytoma from other malignant adrenal masses: importance of wash-in characteristics on multiphase CECT. Endocr Connect 2019; 8:898-905. [PMID: 31252396 PMCID: PMC6599213 DOI: 10.1530/ec-19-0198] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022]
Abstract
RATIONALE AND INTRODUCTION To evaluate the computerised tomography (CT) characteristics of phaeochromocytoma (PCC) that differentiate them from other non-benign adrenal masses such as adrenocortical carcinoma (ACC), primary adrenal lymphoma (PAL) and adrenal metastases (AM). METHODS This retrospective study was conducted at a tertiary health care institute from Western India. Patients presented between January 2013 and August 2016 with histological diagnosis of PCC or other non-benign adrenal mass having adequate reviewable imaging data comprising all four CECT phases were included. RESULTS The study cohort consisted of 72 adrenal masses from 66 patients (33 PCC, 22 ACC, 4 PAL, 13 AM). Unlike other masses, majority of PCC (25/33) showed peak enhancement in early arterial phase (EAP). PCC had significantly higher attenuation in EAP and early venous phase (EVP), and higher calculated percentage arterial enhancement (PAE) and percentage venous enhancement (PVE) than other adrenal masses (P < 0.001). For diagnosis of PCC with 100% specificity, PAE value ≥100% and EAP attenuation ≥100 HU had 78.8 and 63.6% sensitivity respectively. ACC were significantly larger in size as compared to PCC and metastasis. The adreniform shape was exclusively found in PAL (two out of four) and AM (4 out of 13). None of the enhancement, wash-in or washout characteristics were discriminatory among ACC, PAL and AM. CONCLUSION Peak enhancement in EAP, PAE value ≥100% and EAP attenuation ≥100 HU differentiate PCC from other malignant adrenal masses with high specificity.
Collapse
Affiliation(s)
| | - Swati S Jadhav
- Jawaharlal Nehru Medical College, Belagavi, India
- Correspondence should be addressed to S S Jadhav:
| | - Vijaya Sarathi
- Narayana Medical College and Hospital, Nellore, Andhra Pradesh, India
| | | | | | | | - Priya Hira
- Department of Radiology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Rajaram Sharma
- Department of Radiology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Shettepppa Goroshi
- Department of Radiology, Sri Nijalingappa Medical College Bagalkot, Navanagar, Karnataka, India
| | - Gwendolyn Fernandes
- Department of Pathology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Amey Rojekar
- Department of Pathology, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Abhay Dalvi
- Departments of General Surgery, Seth GS Medical College and KEM Hospital, Parel, Mumbai, India
| | - Ganesh Bakshi
- Department of Uro-Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Gagan Prakash
- Department of Uro-Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | | | | |
Collapse
|
21
|
Suh YJ, Park JH, Bilegsaikhan SE, Lee DJ. Transcriptome Analysis Reveals Significant Differences in Gene Expression of Malignant Pheochromocytoma or Paraganglioma. Int J Endocrinol 2019; 2019:7014240. [PMID: 31205467 PMCID: PMC6530119 DOI: 10.1155/2019/7014240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/13/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022] Open
Abstract
Prediction of malignant behavior of pheochromocytoma (PC) or paraganglioma (PG) is of limited value. The Cancer Genome Atlas (TCGA) and the French 'Cortico et Médullosurrénale: les Tumeurs Endocrines' (COMETE) network in Paris (France) facilitate accurate differentiation of malignant PC/PG based on genetic information. Therefore, the objective of this transcriptome analysis is to identify the prognostic genes underlying the differentiation of malignant PC/PG in the TCGA and COMETE databases. TCGA carries data pertaining to multigenomic analysis of 173 PC/PG surgical resection samples while the COMETE cohort contains data involving 188 PC/PG surgical resection samples. Clinical information and mRNA expression datasets were downloaded from TCGA and COMETE databases. Based on eligibility criteria, 58 of 173 PC/PG samples in TCGA and 171 of 188 PC/PG samples collected by the COMETE network were selected. Using Ingenuity Pathway Analysis, the mRNA expression of malignant and benign PC/PG was compared. The 58 samples in TCGA included 11 malignant and 47 benign cases. Among the 171 samples obtained from the COMETE cohort, 19 were malignant and 152 were benign. A comparative analysis of the mRNA expression data of the two databases revealed that 11 up/downregulated pathways involved in malignant PC/PG were related to cancer signaling, metabolic alteration, and prominent mitosis, whereas 6 upregulated genes and 1 downregulated gene were significantly enriched in the functional annotation pathways. The TCGA and COMETE databases showed differences in mRNA expression associated with malignant and benign PC/PG. Improved recognition of prognostic genes facilitates the diagnosis and treatment of PC/PG.
Collapse
Affiliation(s)
- Yong Joon Suh
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Jung Ho Park
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Sanchir-Erdene Bilegsaikhan
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Dong Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| |
Collapse
|
22
|
Cavenagh T, Patel J, Nakhla N, Elstob A, Ingram M, Barber B, Snape K, Bano G, Vlahos I. Succinate dehydrogenase mutations: paraganglioma imaging and at-risk population screening. Clin Radiol 2018; 74:169-177. [PMID: 30551795 DOI: 10.1016/j.crad.2018.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/06/2018] [Indexed: 01/17/2023]
Abstract
Paragangliomas are rare vascular tumours of the autonomic nervous system. They can be classified as sympathetic or parasympathetic. Sympathetic paragangliomas, which include phaeochromocytomas, tend to be functional and symptomatic. Parasympathetic paragangliomas are usually non-functional and may present with mass effect. Forty percent of paragangliomas are linked to genetic syndromes, most commonly due to mutations of the succinate dehydrogenase (SDH) enzyme complex and are collectively known as paraganglioma syndromes, of which five are described. Genetic testing is recommended for all patients, and their first-degree relatives, diagnosed with paragangliomas. When SDH mutations are discovered, biochemical screening and imaging surveillance is indicated. There is currently no consensus on imaging surveillance protocols. Most advocate full-body imaging, but the choice of technique and frequency varies. If paragangliomas are demonstrated, functional imaging to look for synchronous tumours or metastases is indicated. 2-[18F]-fluoro-2-deoxy-d-glucose (18F-FDG) positron-emission tomography (PET)-computed tomography (CT) is the technique of choice for metastatic evaluation, but [123I]-metaiodobenzylguanidine or [111In]-DTPA-octreotide scintigraphy are also utilised. Current research into emerging positron-emitting radiolabelled somatostatin analogues have yielded promising results, which is likely to be reflected in future guidelines. As genetic testing becomes increasingly prevalent, the need to answer the remaining questions regarding surveillance imaging is paramount.
Collapse
Affiliation(s)
- T Cavenagh
- Department of Radiology, St George's University Hospitals NHS Foundation Trust, UK.
| | - J Patel
- Department of Radiology, St George's University Hospitals NHS Foundation Trust, UK
| | - N Nakhla
- Department of Radiology, St George's University Hospitals NHS Foundation Trust, UK
| | - A Elstob
- Department of Radiology, St George's University Hospitals NHS Foundation Trust, UK
| | - M Ingram
- Department of Radiology, Royal Surrey County Hospital, UK
| | - B Barber
- Department of Radiology, Frimley Health NHS Foundation Trust, UK
| | - K Snape
- Department of Medical Genetics, St George's University Hospitals NHS Foundation Trust, UK
| | - G Bano
- Department of Cellular and Molecular Medicine, St George's University Hospitals NHS Foundation Trust, UK
| | - I Vlahos
- Department of Radiology, St George's University Hospitals NHS Foundation Trust, UK
| |
Collapse
|
23
|
Pheochromocytoma in Congenital Cyanotic Heart Disease. Case Rep Endocrinol 2018; 2018:2091257. [PMID: 30356369 PMCID: PMC6176301 DOI: 10.1155/2018/2091257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/03/2018] [Accepted: 09/11/2018] [Indexed: 11/17/2022] Open
Abstract
Studies on genome-wide transcription patterns have shown that many genetic alterations implicated in pheochromocytoma-paraganglioma (P-PGL) syndromes cluster in a common cellular pathway leading to aberrant activation of molecular response to hypoxia in normoxic conditions (the pseudohypoxia hypothesis). Several cases of P-PGL have been reported in patients with cyanotic congenital heart disease (CCHD). Patients affected with CCHD have an increased likelihood of P-PGL compared to those affected with noncyanotic congenital heart disease. One widely supported hypothesis is that chronic hypoxia represents the determining factor supporting this increased risk. We report the case of a 23-year-old woman affected with congenital tricuspid atresia surgically by the Fontan procedure. The patient was admitted to hospital with hypertensive crisis and dyspnea. Chest computed tomography revealed, incidentally, a 6-cm mass in the left adrenal lodge. Increased levels of noradrenaline (NA) and its metabolites were detected (plasma NA 5003.7 pg/ml, n.v.<480; urinary NA 1059.5 µg/24 h, n.v.<85.5; urinary metanephrine 489 µg/24 h, n.v.<320). The patient did not report any additional symptom related to catecholamine excess. The left adrenal tumor showed abnormal accumulation when 131I-metaiodobenzylguanidine scintigraphy was performed. A 18F-fluorodeoxyglucose positron emission tomography showed no significant metabolic activity in the left adrenal gland but intense uptake in the supra- and subdiaphragmatic brown adipose tissue, probably due to noradrenergic-stimulated glucose uptake. The patient underwent left open adrenalectomy after preconditioning with α- and β-blockers and histopathological examination confirmed the diagnosis of pheochromocytoma (Ki-67<5%). Screening for germline mutations did not show any genes mutation (investigated mutations: RET, TMEM127, MAX, SDHD, SDHC, SDHB, SDHAF2, SDHA, and VHL). Clinicians should consider P-PGL when an unexplained clinical deterioration occurs in CCHD patients, even in the absence of typical paroxysmal symptoms.
Collapse
|
24
|
Yamamoto M, Inohara H, Nakagawa T. Targeting metabolic pathways for head and neck cancers therapeutics. Cancer Metastasis Rev 2018; 36:503-514. [PMID: 28819926 DOI: 10.1007/s10555-017-9691-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer cells have distinctive energy metabolism pathways that support their rapid cell division. The preference for anaerobic glycolysis under the normal oxygen condition is known as the Warburg effect and has been observed in head and neck cancers. These metabolic changes are controlled by cancer-related transcription factors, such as tumor suppressor gene and hypoxia inducible factor 1α. In addition, various metabolic enzymes also actively regulate cancer-specific metabolism including the switch between aerobic and anaerobic glycolysis. For a long time, these metabolic changes in cancer cells have been considered a consequence of transformation required to maintain the high rate of tumor cell replication. However, recent studies indicate that alteration of metabolism is sufficient to initiate tumor transformation. Indeed, oncogenic mutations in the metabolic enzymes, isocitrate dehydrogenase and succinate dehydrogenase, have been increasingly found in various cancers, including head and neck cancers. In the present review, we introduce recent findings regarding the cancer metabolism, including the molecular mechanisms of how they affect cancer pathogenesis and maintenance. We also discuss the current and future perspectives on therapeutics that target metabolic pathways, with an emphasis on head and neck cancer.
Collapse
Affiliation(s)
- Masashi Yamamoto
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan.,Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan. .,Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
25
|
Jochmanova I, Pacak K. Pheochromocytoma: The First Metabolic Endocrine Cancer. Clin Cancer Res 2018; 22:5001-5011. [PMID: 27742786 DOI: 10.1158/1078-0432.ccr-16-0606] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/23/2016] [Indexed: 01/21/2023]
Abstract
Dysregulated metabolism is one of the key characteristics of cancer cells. The most prominent alterations are present during regulation of cell respiration, which leads to a switch from oxidative phosphorylation to aerobic glycolysis. This metabolic shift results in activation of numerous signaling and metabolic pathways supporting cell proliferation and survival. Recent progress in genetics and metabolomics has allowed us to take a closer look at the metabolic changes present in pheochromocytomas (PHEO) and paragangliomas (PGL). These neuroendocrine tumors often exhibit dysregulation of mitochondrial metabolism, which is driven by mutations in genes encoding Krebs cycle enzymes or by activation of hypoxia signaling. Present metabolic changes are involved in processes associated with tumorigenesis, invasiveness, metastasis, and resistance to various cancer therapies. In this review, we discuss the metabolic nature of PHEOs/PGLs and how unveiling the metabolic disturbances present in tumors could lead to identification of new biomarkers and personalized cancer therapies. Clin Cancer Res; 22(20); 5001-11. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "ENDOCRINE CANCERS REVISING PARADIGMS".
Collapse
Affiliation(s)
- Ivana Jochmanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland. First Department of Internal Medicine, Medical Faculty of P.J. Šafárik University in Košice, Košice, Slovakia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.
| |
Collapse
|
26
|
Tabebi M, Söderkvist P, Jensen LD. Hypoxia Signaling and Circadian Disruption in and by Pheochromocytoma. Front Endocrinol (Lausanne) 2018; 9:612. [PMID: 30386298 PMCID: PMC6198511 DOI: 10.3389/fendo.2018.00612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/26/2018] [Indexed: 12/30/2022] Open
Abstract
Disruption of the daily (i.e., circadian) rhythms of cell metabolism, proliferation and blood perfusion is a hallmark of many cancer types, perhaps most clearly exemplified by the rare but detrimental pheochromocytomas. These tumors arise from genetic disruption of genes critical for hypoxia signaling, such as von Hippel-Lindau and hypoxia-inducible factor-2 or cellular metabolism, such as succinate dehydrogenase, which in turn impacts on the cellular circadian clock function by interfering with the Bmal1 and/or Clock transcription factors. While pheochromocytomas are often non-malignant, the resulting changes in cellular physiology are coupled to de-regulated production of catecholamines, which in turn disrupt circadian blood pressure variation and therefore circadian entrainment of other tissues. In this review we thoroughly discuss the molecular and physiological interplay between hypoxia signaling and the circadian clock in pheochromocytoma, and how this underlies endocrine disruption leading to loss of circadian blood pressure variation in the affected patients. We furthermore discuss potential avenues for targeting these tumor-specific pathophysiological mechanisms therapeutically in the future.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Peter Söderkvist
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lasse D. Jensen
- Department of Medicine and Health Science, Linköping University, Linköping, Sweden
- *Correspondence: Lasse D. Jensen
| |
Collapse
|
27
|
Zhao B, Zhou Y, Zhao Y, Zhao Y, Wu X, Bi Y, Luo Y, Ji Z, Rong S. Co-Occurrence of Pheochromocytoma-Paraganglioma and Cyanotic Congenital Heart Disease: A Case Report and Literature Review. Front Endocrinol (Lausanne) 2018; 9:165. [PMID: 29719528 PMCID: PMC5914282 DOI: 10.3389/fendo.2018.00165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/28/2018] [Indexed: 11/20/2022] Open
Abstract
Pheochromocytoma and paraganglioma (PHEO-PGL) and cyanotic congenital heart disease (CCHD) are both rare diseases. We reported a 30-year-old patient with a right adrenal gland nodule and a retroperitoneal mass and history of functional single atrium and ventricle. 123I-metaiodobenzylguanidine scintigraphy showed intense uptake in both lesions. Laboratory investigation demonstrated elevated urinary norepinephrine. Preoperative α-blockade was initiated. A successful open resection of right adrenal and retroperitoneal masses was performed. Pathological examination confirmed PHEO-PGL. Postoperative urinary norepinephrine returned to normal level. A systematic case review in English publications in PubMed and EMBASE suggested a hypothesis that there may exist a possible link between PHEO-PGL and hypoxia from CCHD, which was also indicated in our case. Due to higher risk for PHEO-PGL, a lower threshold of suspicion should be considered in CCHD patients. Therefore, active screening and early treatment of PHEO-PGL are recommended in CCHD patients and clinicians should keep on a long-term follow-up to monitor PHEO-PGL recurrence if hypoxia is not corrected.
Collapse
Affiliation(s)
- Bingbin Zhao
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Zhou
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Zhao
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yumo Zhao
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xingcheng Wu
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yalan Bi
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yufeng Luo
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shi Rong
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Shi Rong,
| |
Collapse
|
28
|
Diagnosis and Management of Noncardiac Complications in Adults With Congenital Heart Disease: A Scientific Statement From the American Heart Association. Circulation 2017; 136:e348-e392. [DOI: 10.1161/cir.0000000000000535] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Life expectancy and quality of life for those born with congenital heart disease (CHD) have greatly improved over the past 3 decades. While representing a great advance for these patients, who have been able to move from childhood to successful adult lives in increasing numbers, this development has resulted in an epidemiological shift and a generation of patients who are at risk of developing chronic multisystem disease in adulthood. Noncardiac complications significantly contribute to the morbidity and mortality of adults with CHD. Reduced survival has been documented in patients with CHD with renal dysfunction, restrictive lung disease, anemia, and cirrhosis. Furthermore, as this population ages, atherosclerotic cardiovascular disease and its risk factors are becoming increasingly prevalent. Disorders of psychosocial and cognitive development are key factors affecting the quality of life of these individuals. It is incumbent on physicians who care for patients with CHD to be mindful of the effects that disease of organs other than the heart may have on the well-being of adults with CHD. Further research is needed to understand how these noncardiac complications may affect the long-term outcome in these patients and what modifiable factors can be targeted for preventive intervention.
Collapse
|
29
|
Kavinga Gunawardane PT, Grossman A. The clinical genetics of phaeochromocytoma and paraganglioma. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2017; 61:490-500. [PMID: 29166454 PMCID: PMC10522248 DOI: 10.1590/2359-3997000000299] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 09/26/2017] [Indexed: 11/22/2022]
Abstract
Phaeochromocytoma and paraganglioma are rare catecholamine-producing tumours, recognised to have one of the richest hereditary backgrounds of all neoplasms, with germline mutations seen in approximately 30% of patients. They can be a part of genetic syndromes such as MEN 2 or Neurofibromatosis type 1, or can be found as apparently sporadic tumours. Germline mutations are almost always found in syndromic patients. Nonetheless, apparently sporadic phaeochromocytoma too show high germline mutation rates. Early detection of a genetic mutation can lead to early diagnosis of further tumours via surveillance, early treatment and better prognosis. Apart from this, the genetic profile has important relevance for tumour location and biochemical profile, and can be a useful predictor of future tumour behaviour. It also enables family screening and surveillance. Moreover, recent studies have demonstrated significant driver somatic mutations in up to 75% of all tumours. Arch Endocrinol Metab. 2017;61(5):490-500.
Collapse
Affiliation(s)
- P. T. Kavinga Gunawardane
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordUKOxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, UK
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordUKOxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, UK
- Green Templeton CollegeUniversity of OxfordUKGreen Templeton College, University of Oxford, UK
| |
Collapse
|
30
|
Pang Y, Yang C, Schovanek J, Wang H, Bullova P, Caisova V, Gupta G, Wolf KI, Semenza GL, Zhuang Z, Pacak K. Anthracyclines suppress pheochromocytoma cell characteristics, including metastasis, through inhibition of the hypoxia signaling pathway. Oncotarget 2017; 8:22313-22324. [PMID: 28423608 PMCID: PMC5410225 DOI: 10.18632/oncotarget.16224] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/03/2017] [Indexed: 01/08/2023] Open
Abstract
Pheochromocytomas (PHEOs) and paragangliomas (PGLs) are rare, neuroendocrine tumors derived from adrenal or extra-adrenal chromaffin cells, respectively. Metastases are discovered in 3-36% of patients at the time of diagnosis. Currently, only suboptimal treatment options exist. Therefore, new therapeutic compounds targeting metastatic PHEOs/PGLs are urgently needed. Here, we investigated if anthracyclines were able to suppress the progression of metastatic PHEO. We explored their effects on experimental mouse PHEO tumor cells using in vitro and in vivo models, and demonstrated that anthracyclines, particularly idarubicin (IDA), suppressed hypoxia signaling by preventing the binding of hypoxia-inducible factor 1 and 2 (HIF-1 and HIF-2) to the hypoxia response element (HRE) sites on DNA. This resulted in reduced transcriptional activation of HIF target genes, including erythropoietin (EPO), phosphoglycerate kinase 1 (PGK1), endothelin 1 (EDN1), glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), and vascular endothelial growth factor (VEGFA), which consequently inhibited the growth of metastatic PHEO. Additionally, IDA downregulated hypoxia signaling by interfering with the transcriptional activation of HIF1A and HIF2A. Furthermore, our animal model demonstrated the dose-dependent suppressive effect of IDA on metastatic PHEO growth in vivo. Our results indicate that anthracyclines are prospective candidates for inclusion in metastatic PHEO/PGL therapy, especially in patients with gene mutations involved in the hypoxia signaling pathway.
Collapse
Affiliation(s)
- Ying Pang
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jan Schovanek
- Department of Internal Medicine III-Nephrology, Rheumatology, and Endocrinology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Herui Wang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Petra Bullova
- Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Garima Gupta
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine I Wolf
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Gregg L Semenza
- McKusick-Nathans Institute of Genetic Medicine and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Zhikrivetskaya SO, Snezhkina AV, Zaretsky AR, Alekseev BY, Pokrovsky AV, Golovyuk AL, Melnikova NV, Stepanov OA, Kalinin DV, Moskalev AA, Krasnov GS, Dmitriev AA, Kudryavtseva AV. Molecular markers of paragangliomas/pheochromocytomas. Oncotarget 2017; 8:25756-25782. [PMID: 28187001 PMCID: PMC5421967 DOI: 10.18632/oncotarget.15201] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Paragangliomas/pheochromocytomas comprise rare tumors that arise from the extra-adrenal paraganglia, with an incidence of about 2 to 8 per million people each year. Approximately 40% of cases are due to genetic mutations in at least one out of more than 30 causative genes. About 25-30% of pheochromocytomas/paragangliomas develop under the conditions of a hereditary tumor syndrome a third of which are caused by mutations in the VHL gene. Together, the gene mutations in this disorder have implicated multiple processes including signaling pathways, translation initiation, hypoxia regulation, protein synthesis, differentiation, survival, proliferation, and cell growth. The present review contemplates the mutations associated with the development of pheochromocytomas/paragangliomas and their potential to serve as specific markers of these tumors and their progression. These data will improve our understanding of the pathogenesis of these tumors and likely reveal certain features that may be useful for early diagnostics, malignancy prognostics, and the determination of new targets for disease therapeutics.
Collapse
Affiliation(s)
| | | | - Andrew R Zaretsky
- M.M. Shemyakin - Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Oleg A Stepanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
32
|
Pamporaki C, Hamplova B, Peitzsch M, Prejbisz A, Beuschlein F, Timmers HJ, Fassnacht M, Klink B, Lodish M, Stratakis CA, Huebner A, Fliedner S, Robledo M, Sinnott RO, Januszewicz A, Pacak K, Eisenhofer G. Characteristics of Pediatric vs Adult Pheochromocytomas and Paragangliomas. J Clin Endocrinol Metab 2017; 102:1122-1132. [PMID: 28324046 PMCID: PMC5460722 DOI: 10.1210/jc.2016-3829] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/26/2017] [Indexed: 12/22/2022]
Abstract
CONTEXT Pheochromocytomas and paragangliomas (PPGLs) in children are often hereditary and may present with different characteristics compared with adults. Hereditary PPGLs can be separated into cluster 1 and cluster 2 tumors due to mutations impacting hypoxia and kinase receptor signaling pathways, respectively. OBJECTIVE To identify differences in presentation of PPGLs between children and adults. DESIGN A retrospective cross-sectional clinical study. SETTING Seven tertiary medical centers. PATIENTS The study included 748 patients with PPGLs, including 95 with a first presentation during childhood. Genetic testing was available in 611 patients. Other data included locations of primary tumors, presence of recurrent or metastatic disease, and plasma concentrations of metanephrines and 3-methoxytyramine. RESULTS Children showed higher (P < 0.0001) prevalence than adults of hereditary (80.4% vs 52.6%), extra-adrenal (66.3% vs 35.1%), multifocal (32.6% vs 13.5%), metastatic (49.5% vs 29.1%), and recurrent (29.5% vs 14.2%) PPGLs. Tumors due to cluster 1 mutations were more prevalent among children than adults (76.1% vs 39.3%; P < 0.0001), and this paralleled a higher prevalence of noradrenergic tumors, characterized by relative lack of increased plasma metanephrine, in children than in adults (93.2% vs 57.3%; P < 0.0001). CONCLUSIONS The higher prevalence of hereditary, extra-adrenal, multifocal, and metastatic PPGLs in children than adults represents interrelated features that, in part, reflect the lower age of disease presentation of noradrenergic cluster 1 than adrenergic cluster 2 tumors. The differences in disease presentation are important to consider in children at risk for PPGLs due to a known mutation or previous history of tumor.
Collapse
Affiliation(s)
| | - Barbora Hamplova
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-2425
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the TU Dresden, D-01307 Dresden, Germany
| | | | - Felix Beuschlein
- Department of Medicine IV, University Hospital of Munich, 80539 Munich, Germany
| | - Henri J.L.M. Timmers
- Department of Internal Medicine, Radboud University Medical Centre, 6525 HP Nijmegen, The Netherlands
| | - Martin Fassnacht
- Department of Internal Medicine, Division of Endocrinology, University Hospital, University of Wuerzburg, 97070 Wuerzburg, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus at the TU Dresden, D-01307 Dresden, Germany
- German Cancer Consortium, D-01307 Dresden, Germany
- German Cancer Research Center, 69120 Heidelberg, Germany
- National Center for Tumor Diseases, D-01307 Dresden, Germany
| | - Maya Lodish
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-2425
| | - Constantine A. Stratakis
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-2425
| | | | - Stephanie Fliedner
- Department of Medicine, University Medical Center Schleswig-Holstein, 23562 Luebeck, Germany
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, 28029 Madrid, Spain
| | - Richard O. Sinnott
- Department of Computing and Information, University of Melbourne, 3010 Melbourne, Australia
| | | | - Karel Pacak
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-2425
| | - Graeme Eisenhofer
- Medicine ΙΙI and
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the TU Dresden, D-01307 Dresden, Germany
| |
Collapse
|
33
|
Björklund P, Pacak K, Crona J. Precision medicine in pheochromocytoma and paraganglioma: current and future concepts. J Intern Med 2016; 280:559-573. [PMID: 27165774 PMCID: PMC7441825 DOI: 10.1111/joim.12507] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pheochromocytoma and paraganglioma (PPGL) are rare diseases but are also amongst the most characterized tumour types. Hence, patients with PPGL have greatly benefited from precision medicine for more than two decades. According to current molecular biology and genetics-based taxonomy, PPGL can be divided into three different clusters characterized by: Krebs cycle reprogramming with oncometabolite accumulation or depletion (group 1a); activation of the (pseudo)hypoxia signalling pathway with increased tumour cell proliferation, invasiveness and migration (group 1b); and aberrant kinase signalling causing a pro-mitogenic and anti-apoptotic state (group 2). Categorization into these clusters is highly dependent on mutation subtypes. At least 12 different syndromes with distinct genetic causes, phenotypes and outcomes have been described. Genetic screening tests have a documented benefit, as different PPGL syndromes require specific approaches for optimal diagnosis and localization of various syndrome-related tumours. Genotype-tailored treatment options, follow-up and preventive care are being investigated. Future new developments in precision medicine for PPGL will mainly focus on further identification of driver mechanisms behind both disease initiation and malignant progression. Identification of novel druggable targets and prospective validation of treatment options are eagerly awaited. To achieve these goals, we predict that collaborative large-scale studies will be needed: Pheochromocytoma may provide an example for developing precision medicine in orphan diseases that could ultimately aid in similar efforts for other rare conditions.
Collapse
Affiliation(s)
- P Björklund
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - K Pacak
- Section on Medical Neuroendocrinology, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - J Crona
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
34
|
Tapia-Orihuela RKA, Huaringa-Marcelo J, Loja-Oropeza D. Tetralogy of Fallot and pheochromocytoma in a situs inversus totalis: An unusual association. J Cardiovasc Thorac Res 2016; 8:132-136. [PMID: 27777699 PMCID: PMC5075362 DOI: 10.15171/jcvtr.2016.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/28/2016] [Indexed: 11/22/2022] Open
Abstract
Introduction: Situs inversus totalis is an uncommon anomaly which exist a complete transposition of organs and it’s occasionally associated with congenital heart diseases, such as tetralogy of fallot. Pheochromocytoma is a rare neuroendocrine tumor with an annual incidence of 2-8 cases per million people and for years has been studied its relationship with the hypoxic pathway.
Case Report: A 29 year old male with a history of tetralogy of fallot corrected at 10 years and situs inversus totalis. He was admitted to hospital with a progressive story of four months of constipation, palpitations, headache, dyspnea and sweating. Physical examination revealed a thinned man with peripheral cyanosis, clubbing and signs of decompensated congestive heart failure as hepatomegaly, legs edema, multifocal systodiastolic murmurs, abdominal distension and jugular venous distention. The echocardiogram shows severe right ventricular dysfunction and severe pulmonary hypertension. Furthermore, abdominal computed tomography shows right adrenal mass. Elevated metanephrines and catecholamines confirmed the diagnosis of pheochromocytoma. Surgical removal is decided and preoperative management begins with alpha-adrenergic blockade, however the patient had a hemodynamic decompensation with an unfavorable evolution.
Discussion: In conclusion, there are few reports of cyanotic congenital heart disease with pheochromocytoma. Several studies show a significant association between both of them due to chronic hypoxia leads sustained hyperresponsiveness in adrenal medulla and it would cause the tumor. Special preoperative management of pheochromocytoma is recommended when there underlying heart disease and congestive heart failure. We present the first international report of tetralogy of fallot and pheochromocytoma in a patient with situs inversus totalis.
Collapse
Affiliation(s)
- Rubén Kevin Arnold Tapia-Orihuela
- Universidad Nacional Mayor de San Marcos, Facultad de Medicina de San Fernando, Lima, Perú ; Sociedad Científica de San Fernando, Lima, Perú
| | - Jorge Huaringa-Marcelo
- Universidad Nacional Mayor de San Marcos, Facultad de Medicina de San Fernando, Lima, Perú ; Hospital Nacional Arzobispo Loayza, Lima, Perú
| | - David Loja-Oropeza
- Hospital Nacional Arzobispo Loayza, Lima, Perú ; Universidad Nacional Federico Villarreal, Facultad de Medicina, Lima, Perú
| |
Collapse
|
35
|
Yamamoto K, Namba N, Kubota T, Usui T, Takahashi K, Kitaoka T, Fujiwara M, Hori Y, Kogaki S, Oue T, Morii E, Ozono K. Pheochromocytoma complicated by cyanotic congenital heart disease: a case report. Clin Pediatr Endocrinol 2016; 25:59-65. [PMID: 27212797 PMCID: PMC4860516 DOI: 10.1297/cpe.25.59] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 01/16/2016] [Indexed: 11/04/2022] Open
Abstract
Coincidental cyanotic congenital heart disease and pheochromocytoma is uncommon, although some cases have been reported. We describe a girl aged 15 yr and 11 mo with pheochromocytoma and tricuspid atresia treated by performing the Fontan surgery. The patient did not have any specific symptoms of syndrome related to pheochromoytoma or a family history of pheochromocytoma. During cardiac catheterization, her blood pressure increased markedly, and an α-blocker was administered. Catecholamine hypersecretion was observed in the blood and urine, and abdominal computed tomography revealed a tumor in the right adrenal gland. Scintigraphy showed marked accumulation of (123)I-metaiodobenzylguanidine in the tumor, which led to a diagnosis of pheochromocytoma. We did not detect any germline mutations in the RET, VHL, SDHB, SDHD, TMEM127, or MAX genes. This patient had experienced mild systemic hypoxia since birth, which may have contributed to the development of pheochromocytoma.
Collapse
Affiliation(s)
- Keiko Yamamoto
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Noriyuki Namba
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Pediatrics, Japan Community Health Care Organization, Osaka Hospital, Osaka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Usui
- Clinical Research Institute, National Hospital Organization, Kyoto Medical Center, Kyoto, Japan
| | - Kunihiko Takahashi
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taichi Kitaoka
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yumiko Hori
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigetoyo Kogaki
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takaharu Oue
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
36
|
Pillai S, Gopalan V, Smith RA, Lam AKY. Updates on the genetics and the clinical impacts on phaeochromocytoma and paraganglioma in the new era. Crit Rev Oncol Hematol 2016; 100:190-208. [DOI: 10.1016/j.critrevonc.2016.01.022] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/13/2015] [Accepted: 01/20/2016] [Indexed: 12/18/2022] Open
|
37
|
Du J, Tong A, Wang F, Cui Y, Li C, Zhang Y, Yan Z. The Roles of PI3K/AKT/mTOR and MAPK/ERK Signaling Pathways in Human Pheochromocytomas. Int J Endocrinol 2016; 2016:5286972. [PMID: 27990160 PMCID: PMC5136400 DOI: 10.1155/2016/5286972] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 10/02/2016] [Accepted: 10/27/2016] [Indexed: 11/17/2022] Open
Abstract
Objectives. The roles of PI3K/AKT/mTOR and MAPK/ERK pathways involved in the pathogenesis of pheochromocytoma and paraganglioma (PPGL) were demonstrated mostly by in vitro studies with rat or mouse cells and were mainly studied at transcriptional level. This study aimed to investigate the effect of these pathways on the proliferation of human PPGL cells and the activation of these pathways in PPGLs. Methods. Human PPGL cells were treated with sunitinib and inhibitors of PI3K (LY294002), MEK1/2 (U0126), and mTORC1/2 (AZD8055). Cell proliferation was detected by MTT assay. Protein phosphorylation was detected by Western blotting. Results. In most PPGLs, AKT, ERK1/2, and mTOR were activated. LY294002 (10 μM), U0126 (10 μM), AZD8055 (1 μM), and sunitinib (1 μM) inhibited PPGL cell proliferation in ten primary cultures of tissues, including four from patients with gene mutations. MEK1/2 inhibitor decreased mTOR phosphorylation. Inhibition of mTOR reduced phosphorylation of AKT and ERK1/2. Sunitinib inhibited phospho-ERK1/2 and phospho-mTOR. Conclusion. Our study suggested that PI3K/AKT/mTOR and MAPK/ERK signaling pathways play vital roles in human PPGL and are activated in most PPGLs. Inhibiting multiple pathways might be a novel therapeutic approach for PPGLs.
Collapse
Affiliation(s)
- Juan Du
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Anli Tong
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
- *Anli Tong:
| | - Fen Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Yunying Cui
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Chunyan Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhaoli Yan
- Department of Endocrinology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| |
Collapse
|
38
|
Gravel G, Niccoli P, Rohmer V, Moulin G, Borson-Chazot F, Rousset P, Pasco-Papon A, Marcus C, Dubrulle F, Gouya H, Bidault F, Dupas B, Gabrillargues J, Caumont-Prim A, Hernigou A, Gimenez-Roqueplo AP, Halimi P. The value of a rapid contrast-enhanced angio-MRI protocol in the detection of head and neck paragangliomas in SDHx mutations carriers: a retrospective study on behalf of the PGL.EVA investigators*. Eur Radiol 2015; 26:1696-704. [DOI: 10.1007/s00330-015-4024-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/21/2015] [Accepted: 09/09/2015] [Indexed: 11/24/2022]
|
39
|
Else T. 15 YEARS OF PARAGANGLIOMA: Pheochromocytoma, paraganglioma and genetic syndromes: a historical perspective. Endocr Relat Cancer 2015; 22:T147-59. [PMID: 26273101 DOI: 10.1530/erc-15-0221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The last decades have elucidated the genetic basis of pheochromocytoma (PC) and paraganglioma (PGL) (PCPGL)-associated hereditary syndromes. However, the history of these syndromes dates back at least another 150 years. Detailed descriptions by clinicians and pathologists in the 19th and 20th centuries led to the recognition of the PCPGL-associated syndromes von Hippel-Lindau disease, neurofibromatosis type 1, and multiple endocrine neoplasia type 2. In the beginning of the current millennium the molecular basis of the hereditary PGL syndrome was elucidated by the discovery of mutations in genes encoding enzymes of the Krebs cycle, such as succinate dehydrogenase genes (SDHx) and other mutations, causing 'pseudo-hypoxia' signaling. These recent developments also marked a paradigm shift. It reversed the traditional order of genetic research that historically aimed to define the genetic basis of a known hereditary syndrome but now is challenged with defining the full clinical phenotype associated with a newly defined genetic basis. This challenge underscores the importance to learn from medical history, continue providing support for clinical research, and train physicians with regards to their skills to identify patients with PCPGL-associated syndromes to extend our knowledge of the associated phenotype. This historical overview provides details on the history of the paraganglial system and PCPGL-associated syndromes. As such, it hopefully will not only be an interesting reading for the physician with a historical interest but also emphasize the necessity of ongoing astute individual clinical observations and clinical registries to increase our knowledge regarding the full phenotypic spectrum of these conditions.
Collapse
Affiliation(s)
- Tobias Else
- MetabolismEndocrinology and Diabetes, Department of Internal Medicine, University of Michigan, 2560E MSRB2, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5674, USA
| |
Collapse
|
40
|
Park WJ, Brenner O, Kogot-Levin A, Saada A, Merrill AH, Pewzner-Jung Y, Futerman AH. Development of pheochromocytoma in ceramide synthase 2 null mice. Endocr Relat Cancer 2015; 22:623-32. [PMID: 26113602 PMCID: PMC5586043 DOI: 10.1530/erc-15-0058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 12/12/2022]
Abstract
Pheochromocytoma (PCC) and paraganglioma are rare neuroendocrine tumors of the adrenal medulla and sympathetic and parasympathetic paraganglia, for which mutations in ∼15 disease-associated genes have been identified. We now document the role of an additional gene in mice, the ceramide synthase 2 (CerS2) gene. CerS2, one of six mammalian CerS, synthesizes ceramides with very-long (C22-C24) chains. The CerS2 null mouse has been well characterized and displays lesions in several organs including the liver, lung and the brain. We now demonstrate that changes in the sphingolipid acyl chain profile of the adrenal gland lead to the generation of adrenal medullary tumors. Histological analyses revealed that about half of the CerS2 null mice developed PCC by ∼13 months, and the rest showed signs of medullary hyperplasia. Norepinephrine and normetanephrine levels in the urine were elevated at 7 months of age consistent with the morphological abnormalities found at later ages. Accumulation of ceroid in the X-zone was observed as early as 2 months of age and as a consequence, older mice displayed elevated levels of lysosomal cathepsins, reduced proteasome activity and reduced activity of mitochondrial complex IV by 6 months of age. Together, these findings implicate an additional pathway that can lead to PCC formation, which involves alterations in the sphingolipid acyl chain length. Analysis of the role of sphingolipids in PCC may lead to further understanding of the mechanism by which PCC develops, and might implicate the sphingolipid pathway as a possible novel therapeutic target for this rare tumor.
Collapse
Affiliation(s)
- Woo-Jae Park
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Ori Brenner
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Aviram Kogot-Levin
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Ann Saada
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Alfred H Merrill
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Yael Pewzner-Jung
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | - Anthony H Futerman
- Department of Biological ChemistryWeizmann Institute of Science, Rehovot 76100, IsraelDepartment of BiochemistrySchool of Medicine, Gachon University, Incheon 406-799, South KoreaDepartment of Veterinary ResourcesWeizmann Institute of Science, Rehovot 76100, IsraelMonique and Jacques Roboh Department of Genetic ResearchDepartment of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, IsraelSchool of Biology and Petit Institute for Bioengineering and BioscienceGeorgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| |
Collapse
|
41
|
Jochmanová I, Zhuang Z, Pacak K. Pheochromocytoma: Gasping for Air. Discov Oncol 2015; 6:191-205. [PMID: 26138106 DOI: 10.1007/s12672-015-0231-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/19/2015] [Indexed: 02/06/2023] Open
Abstract
There has been increasing evidence that pseudohypoxia--a phenomenon that we refer to as "gasping for air"--along with mitochondrial enzyme dysregulation play a crucial role in tumorigenesis, particularly in several hereditary pheochromocytomas (PHEOs) and paragangliomas (PGLs). Alterations in key tricarboxylic acids (TCA) cycle enzymes (SDH, FH, MDH2) have been shown to induce pseudohypoxia via activation of the hypoxia-inducible transcription factor (HIF) signaling pathway that is involved in tumorigenesis, invasiveness, and metastatic spread, including an association with resistance to various cancer therapies and worse prognosis. This review outlines the ongoing story of the pathogenesis of hereditary PHEOs/PGLs, showing the unique and most updated evidence of TCA cycle dysregulation that is tightly linked to hypoxia signaling.
Collapse
Affiliation(s)
- Ivana Jochmanová
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver NICHD, National Institutes of Health, Building 10, CRC, 1-East, Room 1E-3140, 10 Center Drive, MSC-1109, Bethesda, MD, 20892-1109, USA.,1st Department of Internal Medicine, Medical Faculty, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Karel Pacak
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver NICHD, National Institutes of Health, Building 10, CRC, 1-East, Room 1E-3140, 10 Center Drive, MSC-1109, Bethesda, MD, 20892-1109, USA.
| |
Collapse
|
42
|
Rethinking pheochromocytomas and paragangliomas from a genomic perspective. Oncogene 2015; 35:1080-9. [DOI: 10.1038/onc.2015.172] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 12/12/2022]
|
43
|
Her YF, Nelson-Holte M, Maher LJ. Oxygen concentration controls epigenetic effects in models of familial paraganglioma. PLoS One 2015; 10:e0127471. [PMID: 25985299 PMCID: PMC4436181 DOI: 10.1371/journal.pone.0127471] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/15/2015] [Indexed: 01/14/2023] Open
Abstract
Familial paraganglioma (PGL) is a rare neuroendocrine cancer associated with defects in the genes encoding the subunits of succinate dehydrogenase (SDH), a tricarboxylic acid (TCA) cycle enzyme. For unknown reasons, a higher prevalence of PGL has been reported for humans living at higher altitude, with increased disease aggressiveness and morbidity. In this study, we evaluate the effects of oxygen on epigenetic changes due to succinate accumulation in three SDH loss cell culture models. We test the hypothesis that the mechanism of α-ketoglutarate (α-KG)-dependent dioxygenase enzymes explains the inhibitory synergy of hypoxia and succinate accumulation. We confirm that SDH loss leads to profound succinate accumulation. We further show that hypoxia and succinate accumulation synergistically inhibit α-KG-dependent dioxygenases leading to increased stabilization of transcription factor HIF1α, HIF2α, and hypermethylation of histones and DNA. Increasing oxygen suppresses succinate inhibition of α-KG-dependent dioxygenases. This result provides a possible explanation for the association between hypoxia and PGL, and suggests hyperoxia as a potential novel therapy.
Collapse
Affiliation(s)
- Yeng F. Her
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, United States of America
- Mayo Graduate School, Mayo Medical School and the Mayo Clinic Medical Scientist Training Program, 200 First St. SW, Rochester, MN, 55905, United States of America
| | - Molly Nelson-Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, United States of America
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN, 55905, United States of America
- * E-mail:
| |
Collapse
|
44
|
Opotowsky AR, Moko LE, Ginns J, Rosenbaum M, Greutmann M, Aboulhosn J, Hageman A, Kim Y, Deng LX, Grewal J, Zaidi AN, Almansoori G, Oechslin E, Earing M, Landzberg MJ, Singh MN, Wu F, Vaidya A. Pheochromocytoma and paraganglioma in cyanotic congenital heart disease. J Clin Endocrinol Metab 2015; 100:1325-34. [PMID: 25581599 PMCID: PMC4399286 DOI: 10.1210/jc.2014-3863] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CONTEXT Aberrant cellular oxygen sensing is a leading theory for development of pheochromocytoma (PHEO) and paraganglioma (PGL). OBJECTIVE The objective of the study was to test the hypothesis that chronic hypoxia in patients with cyanotic congenital heart disease (CCHD) increases the risk for PHEO-PGL. DESIGN/SETTING/PARTICIPANTS We investigated the association between CCHD and PHEO-PGL with two complementary studies: study 1) an international consortium was established to identify congenital heart disease (CHD) patients with a PHEO-PGL diagnosis confirmed by pathology or biochemistry and imaging; study 2) the 2000-2009 Nationwide Inpatient Survey, a nationally representative discharge database, was used to determine population-based cross-sectional PHEO-PGL frequency in hospitalized CCHD patients compared with noncyanotic CHD and those without CHD using multivariable logistic regression adjusted for age, sex, and genetic PHEO-PGL syndromes. RESULTS In study 1, we identified 20 PHEO-PGL cases, of which 18 had CCHD. Most presented with cardiovascular or psychiatric symptoms. Median cyanosis duration for the CCHD PHEO-PGL cases was 20 years (range 1-57 y). Cases were young at diagnosis (median 31.5 y, range 15-57 y) and 7 of 18 had multiple tumors (two bilateral PHEO; six multifocal or recurrent PGL), whereas 11 had single tumors (seven PHEO; four PGL). PGLs were abdominal (13 of 17) or head/neck (4 of 17). Cases displayed a noradrenergic biochemical phenotype similar to reported hypoxia-related PHEO-PGL genetic syndromes but without clinical signs of such syndromes. In study 2, hospitalized CCHD patients had an increased likelihood of PHEO-PGL (adjusted odds ratio 6.0, 95% confidence interval 2.6-13.7, P < .0001) compared with those without CHD; patients with noncyanotic CHD had no increased risk (odds ratio 0.9, P = .48). CONCLUSIONS There is a strong link between CCHD and PHEO-PGL. Whether these rare diseases coassociate due to hypoxic stress, common genetic or developmental factors, or some combination requires further investigation.
Collapse
Affiliation(s)
- Alexander R Opotowsky
- Department of Cardiology (A.R.O., L.E.M., M.J.L., M.N.S., F.W.), Boston Children's Hospital, Boston, Massachusetts 02115; Division of Cardiovascular Medicine, (A.R.O., M.J.L., M.N.S., F.W.), Division of Endocrinology (A.V.), Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Medicine (J.Gi., M.R.), Columbia University Medical Center, New York, New York 10027; Adult Congenital Heart Disease Program (M.G.), University Hospital Zurich, CH-8032 Zurich, Switzerland; Department of Medicine (J.A.,A.H.), Division of Cardiology, University of California, Los Angeles, Medical Center, Ahmanson/UCLA Adult Congenital Heart Disease Center, Los Angeles, California 90095; Department of Cardiology (Y.K., L.X.D.), The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Department of Medicine (Y.K., L.X.D.), Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104; Division of Cardiology (J.Gr.), St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4H4; The Heart Center (A.N.Z.), Nationwide Children's Hospital, Columbus, Ohio 43205; Department of Internal Medicine (A.N.Z.), The Ohio State University Wexner Medical Center, Columbus, Ohio 43210; Department of Medicine (G.A., E.O.), University Health Network and University of Toronto, Toronto, Ontario, CanadaM5G2C4; Department of Pediatrics (M.E.), Medical College of Wisconsin, Milwaukee, Wisconsin 53226; Center for Adrenal Disorders (A.V.), Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Jochmanová I, Zelinka T, Widimský J, Pacak K. HIF signaling pathway in pheochromocytoma and other neuroendocrine tumors. Physiol Res 2015; 63:S251-62. [PMID: 24908231 DOI: 10.33549/physiolres.932789] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors controlling energy, iron metabolism, erythropoiesis, and development. Dysregulation of these proteins contributes to tumorigenesis and cancer progression. Recent findings revealed the important role of HIFs in the pathogenesis of neuroendocrine tumors, especially pheochromocytoma (PHEO) and paraganglioma (PGL). PHEOs and PGLs are catecholamine-producing tumors arising from sympathetic- or parasympathetic-derived chromaffin tissue. To date, eighteen PHEO/PGL susceptibility genes have been identified. Based on the main signaling pathways, PHEOs/PGLs have been divided into two clusters, pseudohypoxic cluster 1 and cluster 2, rich in kinase receptor signaling and protein translation pathways. Recent data suggest that both clusters are interconnected via the HIF signaling and its role in tumorigenesis is supported by newly described somatic and germline mutations in HIF2A gene in patients with PHEOs/PGLs associated with polycythemia, and in some of them also with somatostatinoma. Moreover, HIFalpha signaling has also been shown to be upregulated in neuroendocrine tumors other than PHEO/PGL. Some of these tumors are components of hereditary tumor syndromes which can be associated with PHEO/PGL, but also in ileal carcinoids or melanoma. HIF signaling appears to be one of the crucial players in tumorigenesis, which could suggest new therapeutic approaches for treatment of neuroendocrine tumors.
Collapse
Affiliation(s)
- I Jochmanová
- Department of Internal Medicine, Faculty of Medicine, P. J. Šafárik University, Košice, Slovakia; Section on Medical Neuroendocrinology, Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD, USA.
| | | | | | | |
Collapse
|
46
|
Luchetti A, Walsh D, Rodger F, Clark G, Martin T, Irving R, Sanna M, Yao M, Robledo M, Neumann HPH, Woodward ER, Latif F, Abbs S, Martin H, Maher ER. Profiling of somatic mutations in phaeochromocytoma and paraganglioma by targeted next generation sequencing analysis. Int J Endocrinol 2015; 2015:138573. [PMID: 25883647 PMCID: PMC4390106 DOI: 10.1155/2015/138573] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/23/2014] [Accepted: 08/26/2014] [Indexed: 01/24/2023] Open
Abstract
At least 12 genes (FH, HIF2A, MAX, NF1, RET, SDHA, SDHB, SDHC, SDHD, SDHAF2, TMEM127, and VHL) have been implicated in inherited predisposition to phaeochromocytoma (PCC), paraganglioma (PGL), or head and neck paraganglioma (HNPGL) and a germline mutation may be detected in more than 30% of cases. Knowledge of somatic mutations contributing to PCC/PGL/HNPGL pathogenesis has received less attention though mutations in HRAS, HIF2A, NF1, RET, and VHL have been reported. To further elucidate the role of somatic mutation in PCC/PGL/HNPGL tumourigenesis, we employed a next generation sequencing strategy to analyse "mutation hotspots" in 50 human cancer genes. Mutations were identified for HRAS (c.37G>C; p.G13R and c.182A>G; p.Q61R) in 7.1% (6/85); for BRAF (c.1799T>A; p.V600E) in 1.2% (1/85) of tumours; and for TP53 (c.1010G>A; p.R337H) in 2.35% (2/85) of cases. Twenty-one tumours harboured mutations in inherited PCC/PGL/HNPGL genes and no HRAS, BRAF, or TP53 mutations occurred in this group. Combining our data with previous reports of HRAS mutations in PCC/PGL we find that the mean frequency of HRAS/BRAF mutations in sporadic PCC/PGL is 8.9% (24/269) and in PCC/PGL with an inherited gene mutation 0% (0/148) suggesting that HRAS/BRAF mutations and inherited PCC/PGL genes mutations might be mutually exclusive. We report the first evidence for BRAF mutations in the pathogenesis of PCC/PGL/HNPGL.
Collapse
Affiliation(s)
- Andrea Luchetti
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Diana Walsh
- Centre for Rare Diseases and Personalised Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - Fay Rodger
- Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Graeme Clark
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Tom Martin
- Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Birmingham B15 2TH, UK
| | - Richard Irving
- Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Birmingham B15 2TH, UK
| | - Mario Sanna
- Department of Otology & Skull Base Surgery, Gruppo Otologico, Via Antonio Emmanueli 42, 29121 Piacenza, Italy
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Via dei Vestini 1, 66100 Chieti, Italy
| | - Masahiro Yao
- Department of Urology, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa 236-0004, Japan
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Hartmut P. H. Neumann
- Section of Preventive Medicine, Department of Nephrology, Albert Ludwigs University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Emma R. Woodward
- Centre for Rare Diseases and Personalised Medicine, University of Birmingham, Birmingham B15 2TT, UK
- Department of Clinical Genetics, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - Farida Latif
- Centre for Rare Diseases and Personalised Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - Stephen Abbs
- Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Howard Martin
- Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Eamonn R. Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- Centre for Rare Diseases and Personalised Medicine, University of Birmingham, Birmingham B15 2TT, UK
- *Eamonn R. Maher:
| |
Collapse
|
47
|
Abstract
Paragangliomas (PGLs) are rare vascular, neuroendocrine tumors of paraganglia, which are associated with either sympathetic tissue in adrenal (pheochromocytomas (PCCs)) and extraadrenal (sympathetic paraganglioma (sPGLs)) locations or parasympathetic tissue of the head and neck paragangliomas (HNPGLs). As HNPGLs are usually benign and most tumors grow slowly, a wait-and-scan policy is often advised. However, their location in the close proximity to cranial nerves and vasculature may result in considerable morbidity due to compression or infiltration of the adjacent structures, necessitating balanced decisions between a wait-and-see policy and active treatment. The main treatment options for HNPGL are surgery and radiotherapy. In contrast to HNPGLs, the majority of sPGL/PCCs produces catecholamines, in advanced cases resulting in typical symptoms and signs such as palpitations, headache, diaphoresis, and hypertension. The state-of-the-art diagnosis and localization of sPGL/PCCs are based on measurement of plasma and/or 24-h urinary excretion of (fractionated) metanephrines and methoxytyramine (MT). sPGL/PCCs can subsequently be localized by anatomical (computed tomography and/or magnetic resonance imaging) and functional imaging studies (123I-metaiodobenzylguanidine-scintigraphy, 111In-pentetreotide scintigraphy, or positron emission tomography with radiolabeled dopamine or dihydroxyphenylalanine). Although most PGL/PCCs are benign, factors such as genetic background, tumor size, tumor location, and high MT levels are associated with higher rates of metastatic disease. Surgery is the only curative treatment. Treatment options for patients with metastatic disease are limited. PGL/PCCs have a strong genetic background, with at least one-third of all cases linked with germline mutations in 11 susceptibility genes. As genetic testing becomes more widely available, the diagnosis of PGL/PCCs will be made earlier due to routine screening of at-risk patients. Early detection of a familial PGL allows early detection of potentially malignant PGLs and early surgical treatment, reducing the complication rates of this operation.
Collapse
Affiliation(s)
- Eleonora P Corssmit
- Department of EndocrinologyLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The NetherlandsDepartment and Division of MedicineAcademic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johannes A Romijn
- Department of EndocrinologyLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The NetherlandsDepartment and Division of MedicineAcademic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Chronic hypoxemia and paraganglioma: a rare case and discussion of potential mechanisms. Am J Med Sci 2014; 348:528-9. [PMID: 25325194 DOI: 10.1097/maj.0000000000000367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Denorme M, Yon L, Roux C, Gonzalez BJ, Baudin E, Anouar Y, Dubessy C. Both sunitinib and sorafenib are effective treatments for pheochromocytoma in a xenograft model. Cancer Lett 2014; 352:236-44. [PMID: 25016061 DOI: 10.1016/j.canlet.2014.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 10/25/2022]
Abstract
Pheochromocytomas and paragangliomas are rare neuroendocrine tumors which develop from chromaffin cells of the adrenal medulla and extra-adrenal sites, leading to excess catecholamine release and hypertension. Many of the tumors are characterized by a high vascularity, suggesting the possible implementation of anti-angiogenic therapies for patients. Here, the efficacy of the tyrosine kinase inhibitors sunitinib and sorafenib was investigated in vivo and in vitro. Oral treatment with either sunitinib or sorafenib (40mg/kg/day) for 14days induced a marked reduction in the volume and weight of PC12 pheochromocytoma cell tumor xenografts in mice. Assessment of tumoral neo-angiogenesis, assessed by morphometric analysis of the vascular network after CD31 immunolabeling, showed that both sunitinib and sorafenib reduced the microvessel area (-85% and -80%, respectively) and length (-80% and -78%, respectively) in treated compared to control tumors. In addition, the number of vessel nodes was significantly lower in treated tumors (-95% and -84%, respectively). Furthermore, cleaved caspase 3 immunolabeling revealed a marked increase in the number of apoptotic cells in tumors from treated animals. Sunitinib and sorafenib could exert a direct effect on PC12 cell viability in vitro. While sunitinib induced a rapid (4h) and pronounced (5-fold) increase in caspase-3/7-dependent apoptosis, sorafenib seems to exert its cytotoxic activity through a different mechanism. Altogether, our data demonstrate that sunitinib and sorafenib have the ability to impair pheochromocytoma development by inhibiting angiogenesis and reducing tumor cell viability. These results strongly suggest that both sunitinib and sorafenib could represent valuable therapeutic tools for pheochromocytoma.
Collapse
Affiliation(s)
- M Denorme
- Institut National de la Santé et de la Recherche Médicale (INSERM), U982, Mont-Saint-Aignan, France; Normandie Univ, Caen, France; University of Rouen, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France
| | - L Yon
- Institut National de la Santé et de la Recherche Médicale (INSERM), U982, Mont-Saint-Aignan, France; Normandie Univ, Caen, France; University of Rouen, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France
| | - C Roux
- Normandie Univ, Caen, France; Haute-Normandie-INSERM ERI28, Rouen, France; University of Rouen, Laboratory of Microvascular Endothelium and Neonate Brain Lesion, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - B J Gonzalez
- Normandie Univ, Caen, France; Haute-Normandie-INSERM ERI28, Rouen, France; University of Rouen, Laboratory of Microvascular Endothelium and Neonate Brain Lesion, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - E Baudin
- Departments of Nuclear Medicine and Endocrine Tumors, Institut Gustave-Roussy, Villejuif, France
| | - Y Anouar
- Institut National de la Santé et de la Recherche Médicale (INSERM), U982, Mont-Saint-Aignan, France; Normandie Univ, Caen, France; University of Rouen, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France.
| | - C Dubessy
- Institut National de la Santé et de la Recherche Médicale (INSERM), U982, Mont-Saint-Aignan, France; Normandie Univ, Caen, France; University of Rouen, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France
| |
Collapse
|
50
|
Vicha A, Taieb D, Pacak K. Current views on cell metabolism in SDHx-related pheochromocytoma and paraganglioma. Endocr Relat Cancer 2014; 21:R261-77. [PMID: 24500761 PMCID: PMC4016161 DOI: 10.1530/erc-13-0398] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Warburg's metabolic hypothesis is based on the assumption that a cancer cell's respiration must be under attack, leading to its damage, in order to obtain increased glycolysis. Although this may not apply to all cancers, there is some evidence proving that primarily abnormally functioning mitochondrial complexes are indeed related to cancer development. Thus, mutations in complex II (succinate dehydrogenase (SDH)) lead to the formation of pheochromocytoma (PHEO)/paraganglioma (PGL). Mutations in one of the SDH genes (SDHx mutations) lead to succinate accumulation associated with very low fumarate levels, increased glutaminolysis, the generation of reactive oxygen species, and pseudohypoxia. This results in significant changes in signaling pathways (many of them dependent on the stabilization of hypoxia-inducible factor), including oxidative phosphorylation, glycolysis, specific expression profiles, as well as genomic instability and increased mutability resulting in tumor development. Although there is currently no very effective therapy for SDHx-related metastatic PHEOs/PGLs, targeting their fundamental metabolic abnormalities may provide a unique opportunity for the development of novel and more effective forms of therapy for these tumors.
Collapse
Affiliation(s)
- Ales Vicha
- Department of Pediatric Hematology and Oncology, 2 Medical School, Charles University and University Hospital Motol, Prague, Czech Republic
| | - David Taieb
- Service Central de Biophysique et de Médecine Nucléaire, CERIMED Centre hospitalo-universitaire Timone, Marseille, France
- Département d’Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Karel Pacak
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health, Bethesda, Maryland, 20892 USA
| |
Collapse
|