1
|
Jiang J, Pang Y, Luo R, Wei Y, Zhang J, Li M, Xu Y, Teng X, Wu H, Guan H, Wu X, Yan C, Zhong D, Deng W, Xu N, Wen Y, Feng Y, Yan B, Wang L, Jiang Y, Ning J, Xu X, Soria MJ, Robledo M, Pacak K, Liu Y, Liu L. Genetics of urinary bladder paragangliomas: a multi-center study of a Chinese cohort. J Endocrinol Invest 2024:10.1007/s40618-024-02509-w. [PMID: 39636472 DOI: 10.1007/s40618-024-02509-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Pheochromocytomas and paragangliomas (PPGLs) exhibit the highest degree of heritability among all human tumors, yet the genetics of urinary bladder paragangliomas (UBPGLs) remains poorly understood. The present study aims to examine the characteristics of a cohort of Chinese patients with UBPGLs, focusing particularly on genetics. METHODS The study included 70 Chinese patients with UBPGLs from 15 centers in China, 240 patients with non-head and neck PGLs (non-HNPGLs) outside the urine bladder, and 16 Caucasian patients with UBPGLs. Tumor DNA samples were sequenced by next generation sequencing. All identified pathogenic variants (PVs) were confirmed by Sanger sequencing. RESULTS Among the 70 Chinese patients, PVs were identified in 38 cases: 23 in cluster 1 A (13 SDHB, 1 SDHD, 1 SDHA, 4 IDH1, 2 SLC25A11, and 2 FH), 4 in cluster 1B (3 EPAS1 and 1 EGLN1), and 11 in cluster 2 genes (7 HRAS, 1 FGFR1, 2 NF1, and 1 H3F3A). Compared with other non-HNPGLs, UBPGLs had more PVs in cluster 1 A genes (32.9% vs. 14.2%, p < 0.001), but fewer in cluster 1B (5.7% vs. 19.2%, p = 0.002) and cluster 2 genes (15.7% vs. 42.5%, p < 0.001). PVs in SDHB (18.6%) was the most common in Chinese patients with UBPGLs, followed by HRAS (10.0%). No PVs was found in 45.7% of all UBPGLs. PVs in HRAS, SLC25A11, EPAS1, and FH were also identified in Caucasians with UBPGLs. CONCLUSION Chinese patients with UBPGLs have a diverse genetic profile. PVs in cluster 1 A genes underlie nearly 1/3 of patients, highlighting the importance of genetic testing. Diverse germline and somatic PVs are also present in Caucasian patients with UBPGLs.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yongbao Wei
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Jing Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Minghao Li
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yitong Xu
- Department of Pathology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, NHC Key Laboratory of Diagnosis and Treatment of Thyroid Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaohong Wu
- Geriatric Medicine Center, Cancer Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Chenyan Yan
- Geriatric Medicine Center, Cancer Center, Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Dewen Zhong
- Department of Urology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Wanglong Deng
- Department of Urology, the First People's Hospital of Chenzhou, Chenzhou, China
| | - Ning Xu
- Department of Urology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yanlin Wen
- Department of Urology, Nanchong Central Hospital, the Second Clinical Hospital of North Sichuan Medical College, Szechwan, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Yan
- Department of Urology, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yazhuo Jiang
- Department of Urology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaowen Xu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Miguel J Soria
- Department of Medical Oncology, European University of Madrid, University Getafe Hospital, Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, Spanish National Cancer Research Center, Madrid, Spain
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, 180 Fenglin Road Xuhui District, Shanghai, 200032, China.
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Monteagudo M, Calsina B, Salazar-Hidalgo ME, Martínez-Montes ÁM, Piñeiro-Yáñez E, Caleiras E, Martín MC, Rodríguez-Perales S, Letón R, Gil E, Buffet A, Burnichon N, Fernández-Sanromán Á, Díaz-Talavera A, Mellid S, Arroba E, Reglero C, Martínez-Puente N, Roncador G, Del Olmo MI, Corrales PJP, Oliveira CL, Álvarez-Escolá C, Gutiérrez MC, López-Fernández A, García NP, Regojo RM, Díaz LR, Laorden NR, Guadarrama OS, Bechmann N, Beuschlein F, Canu L, Eisenhofer G, Fassnacht M, Nölting S, Quinkler M, Rapizzi E, Remde H, Timmers HJ, Favier J, Gimenez-Roqueplo AP, Rodriguez-Antona C, Currás-Freixes M, Al-Shahrour F, Cascón A, Leandro-García LJ, Montero-Conde C, Robledo M. MAML3-fusions modulate vascular and immune tumour microenvironment and confer high metastatic risk in pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2024; 38:101931. [PMID: 39218714 DOI: 10.1016/j.beem.2024.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pheochromocytomas and paragangliomas are rare neuroendocrine tumours. Around 20-25 % of patients develop metastases, for which there is an urgent need of prognostic markers and therapeutic stratification strategies. The presence of a MAML3-fusion is associated with increased metastatic risk, but neither the processes underlying disease progression, nor targetable vulnerabilities have been addressed. We have compiled a cohort of 850 patients, which has shown a 3.65 % fusion prevalence and represents the largest MAML3-positive series reported to date. While MAML3-fusions mainly cause single pheochromocytomas, we also observed somatic post-zygotic events, resulting in multiple tumours in the same patient. MAML3-tumours show increased expression of neuroendocrine-to-mesenchymal transition markers, MYC-targets, and angiogenesis-related genes, leading to a distinct tumour microenvironment with unique vascular and immune profiles. Importantly, our findings have identified MAML3-tumours specific vulnerabilities beyond Wnt-pathway dysregulation, such as a rich vascular network, and overexpression of PD-L1 and CD40, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- María Monteagudo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Bruna Calsina
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Milton E Salazar-Hidalgo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ángel M Martínez-Montes
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Carmen Martín
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rocío Letón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Gil
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alexandre Buffet
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Nelly Burnichon
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Ángel Fernández-Sanromán
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Díaz-Talavera
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Sara Mellid
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ester Arroba
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Reglero
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Natalia Martínez-Puente
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Isabel Del Olmo
- Department of Endocrinology and Nutrition, University Hospital La Fe, Valencia, Spain
| | | | - Cristina Lamas Oliveira
- Department of Endocrinology and Nutrition Albacete University Hospital, SESCAM, Albacete, Spain
| | | | | | | | | | | | - Luis Robles Díaz
- Department of Oncology, 12 de Octubre University Hospital, Madrid, Spain
| | | | | | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden Germany, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV Klinikum der Universität München, Munich, Germany; Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland; LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Letizia Canu
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Graeme Eisenhofer
- Department of Medicine III University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I Division of Endocrinology and Diabetes University Hospital Würzburg University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Svenja Nölting
- Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland
| | - Marcus Quinkler
- Endocrinology in Charlottenburg Stuttgarter Platz 1, Berlin, Germany
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Hanna Remde
- Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Henri J Timmers
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Judith Favier
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Currás-Freixes
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fatima Al-Shahrour
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Luis J Leandro-García
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Montero-Conde
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
| |
Collapse
|
3
|
de Bresser CJM, de Krijger RR. The Molecular Classification of Pheochromocytomas and Paragangliomas: Discovering the Genomic and Immune Landscape of Metastatic Disease. Endocr Pathol 2024; 35:279-292. [PMID: 39466488 DOI: 10.1007/s12022-024-09830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs, together PPGLs) are the most hereditary tumors known. PPGLs were considered benign, but the fourth edition of the World Health Organisation (WHO) classification redefined all PPGLs as malignant neoplasms with variable metastatic potential. The metastatic rate differs based on histopathology, genetic background, size, and location of the tumor. The challenge in predicting metastatic disease lies in the absence of a clear genotype-phenotype correlation among the more than 20 identified genetic driver variants. Recent advances in molecular clustering based on underlying genetic alterations have paved the way for improved cluster-specific personalized treatments. However, despite some clusters demonstrating a higher propensity for metastatic disease, cluster-specific therapies have not yet been widely adopted in clinical practice. Comprehensive genomic profiling and transcriptomic analyses of large PPGL cohorts have identified potential new biomarkers that may influence metastatic potential. It appears that no single biomarker alone can reliably predict metastatic risk; instead, a combination of these biomarkers may be necessary to develop an effective prediction model for metastatic disease. This review evaluates current guidelines and recent genomic and transcriptomic findings, with the aim of accurately identifying novel biomarkers that could contribute to a predictive model for mPPGLs, thereby enhancing patient care and outcomes.
Collapse
Affiliation(s)
- Carolijn J M de Bresser
- Department of Vascular Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| |
Collapse
|
4
|
Jiang J, Liu Y. Recent discoveries of Sino-Caucasian differences in the genetics of phaeochromocytomas and paragangliomas. Best Pract Res Clin Endocrinol Metab 2024; 38:101928. [PMID: 39191630 DOI: 10.1016/j.beem.2024.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) represent the highest degree of heritability of any known tumor types in humans. Previous studies have characterized a dramatic difference between Chinese and European Caucasians with regards to both genetics and clinical features of PPGLs. The proportion of PGLs in Chinese patients was higher than in Caucasians, and the prevalence of metastasis was much lower in Chinese patients. Compared with Caucasians, there were more pathogenic variants (PVs) found in HRAS and FGFR1, but less in NF1 and SDHB. There were less germline PVs found in Chinese patients. Importantly, in Chinese patients, there was a large proportion of PGLs with PVs found in HRAS and FGFR1, mostly with epinephrine-producing capacity. This finding provided solid evidence that genetics (cluster 1 vs. 2), rather than location (PCC vs. PGL), determines the catecholamine-producing phenotype. Besides, the lower prevalence of SDHB partially explained lower occurrence of metastatic lesions in Chinese patients. These findings underscore the importance of considering ethnic differences when evaluating PPGLs and patient outcomes.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, China.
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
5
|
Boehm E, Gill AJ, Clifton-Bligh R, Tothill RW. Recent progress in molecular classification of phaeochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2024; 38:101939. [PMID: 39271378 DOI: 10.1016/j.beem.2024.101939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Phaeochromocytomas (PC) and paragangliomas (PG) are neural crest cancers with high heritability. Recent advances in molecular profiling, including multi-omics and single cell genomics has identified up to seven distinct molecular subtypes. These subtypes are defined by mutations involving hypoxia-inducible factors (HIFs), Krebs cycle, kinase and WNT signalling, but are also defined by chromaffin differentiation states. PCPG have a dominant proangiogenic microenvironment linked to HIF pathway activity and are generally considered "immune cold" tumours with a high number of macrophages. PCPG subtypes can indicate increased metastatic risk but secondary mutations in telomere maintenance genes TERT or ATRX are required to drive the metastatic phenotype. Molecular profiling can identify molecular therapeutic (e.g. RET and EPAS1) and radiopharmaceutical targets while also helping to support variant pathogenicity and familial risk. Molecular profiling and subtyping of PCPG therefore confers the possibility of nuanced prognostication and individual treatment stratification but this still requires large-scale prospective validation.
Collapse
Affiliation(s)
- Emma Boehm
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia; Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Anthony J Gill
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney NSW, Australia
| | - Roderick Clifton-Bligh
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney NSW, Australia.
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Snezhkina AV, Pavlov VS, Krasnov GS, Kalinin DV, Pudova EA, Stolbovskaya OV, Dunshina AV, Fedorova MS, Kudryavtseva AV. Non-Susceptibility Gene Variants in Head and Neck Paragangliomas. Int J Mol Sci 2024; 25:12762. [PMID: 39684472 DOI: 10.3390/ijms252312762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Head and neck paragangliomas (HNPGLs) are rare neoplasms that, along with pheochromocytomas and extra-adrenal paragangliomas, are associated with inherited mutations in at least 12 susceptibility genes in approximately 40% of cases. However, due to the rarity of HNPGLs, only a series of small-scale studies and individual cases have reported mutations in additional genes that may be involved in tumorigenesis. Consequently, numerous disease-causing mutations and genes responsible for the pathogenesis of HNPGLs remain poorly investigated. The aim of this study was to gain a deeper understanding of the genetic basis of HNPGLs by focusing on variants in genes that were not previously identified as well-known drivers. A whole-exome data analysis was conducted on a representative set of 152 HNPGLs. In 30% of the tumors examined, 53 potentially deleterious variants were identified in 36 different genes. The analysis identified pathogenic or likely pathogenic variants in the ARNT, IDH2, L2HGDH, MYH3, PIK3CA, and TERT genes. A functional network analysis of the mutated genes revealed numerous associations and a list of metabolic pathways (e.g., the TCA cycle, carbon metabolism, pyruvate metabolism, etc.) and signaling pathways (e.g., HIF1, PI3K-Akt, FoxO, AMPK, MAPK, etc.) that may play an important role in the development of HNPGLs. The identified range of genetic alterations affecting multiple genes and, potentially, influencing diverse cellular pathways provides an enhanced molecular genetic characterization of HNPGLs.
Collapse
Affiliation(s)
- Anastasiya V Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladislav S Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Dmitry V Kalinin
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Olga V Stolbovskaya
- Department of Human Anatomy, Ulyanovsk State University, 432017 Ulyanovsk, Russia
| | | | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
7
|
Saintilnord WN, Hegazy YA, Chesnutt K, Eckstein M, Cassidy RN, Dhahri H, Bennett RL, Melters DP, Lopes E, Fu Z, Lau K, Chandler DP, Poirier MG, Dalal Y, Licht JD, Fondufe-Mittendorf Y. Aberrant expression of histone H2B variants reshape chromatin and alter oncogenic gene expression programs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624207. [PMID: 39605447 PMCID: PMC11601509 DOI: 10.1101/2024.11.18.624207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Chromatin architecture governs DNA accessibility and gene expression. Thus, any perturbations to chromatin can significantly alter gene expression programs and promote disease. Prior studies demonstrate that every amino acid in a histone is functionally significant, and that even a single amino acid substitution can drive specific cancers. We previously observed that naturally occurring H2B variants are dysregulated during the epithelial to mesenchymal transition (EMT) in bronchial epithelial cells. Naturally occurring H2B variants differ from canonical H2B by only a few amino acids, yet single amino acid changes in other histone variants (e.g., H3.3) can drive cancer. We therefore hypothesized that H2B variants might function like oncohistones, and investigated how they modify chromatin architecture, dynamics, and function. We find that H2B variants are frequently dysregulated in many cancers, and correlate with patient prognosis. Despite high sequence similarity, mutations in each H2B variant tend to occur at specific "hotspots" in cancer. Some H2B variants cause tighter DNA wrapping around nucleosomes, leading to more compact chromatin structures and reduced transcription factor accessibility to nucleosomal DNA. They also altered genome-wide accessibility to oncogenic regulatory elements and genes, with concomitant changes in oncogenic gene expression programs. Although we did not observe changes in cell proliferation or migration in vitro , our Gene Ontology (GO) analyses of ATAC-seq peaks and RNA-seq data indicated significant changes in oncogenic pathways. These findings suggest that H2B variants may influence early-stage, cancer-associated regulatory mechanisms, potentially setting the stage for oncogenesis later on. Thus, H2B variant expression could serve as an early cancer biomarker, and H2B variants might be novel therapeutic targets.
Collapse
|
8
|
Cascón A, Robledo M. Clinical and molecular markers guide the genetics of pheochromocytoma and paraganglioma. Biochim Biophys Acta Rev Cancer 2024; 1879:189141. [PMID: 38908536 DOI: 10.1016/j.bbcan.2024.189141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Over the past two decades, research into the genetic susceptibility behind pheochromocytoma and paraganglioma (PPGL) has surged, ranking them among the most heritable tumors. Massive sequencing combined with careful patient selection has so far identified more than twenty susceptibility genes, leading to an over-detection of variants of unknown significance (VUS) that require precise molecular markers to determine their pathogenic role. Moreover, some PPGL patients remain undiagnosed, possibly due to mutations in regulatory regions of already known genes or mutations in undiscovered genes. Accurate classification of VUS and identification of new genes require well-defined clinical and molecular markers that allow effective genetic diagnosis of most PPGLs.
Collapse
Affiliation(s)
- Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
9
|
Nguyen T, Ordulu Z, Shrestha S, Patel U, Crispen PL, Brown L, Falzarano SM, Ghayee HK, Perdomo Rodriguez JP. Case report: A novel somatic SDHB variant in a patient with bladder paraganglioma. Front Endocrinol (Lausanne) 2024; 15:1386285. [PMID: 38911036 PMCID: PMC11190172 DOI: 10.3389/fendo.2024.1386285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Background Paragangliomas (PGL) are rare neuroendocrine tumors derived from the autonomic nervous system paraganglia. Urinary bladder paragangliomas (UBPGL) originate from the sympathetic neurons of the urinary bladder wall and represent 0.7% of all paragangliomas and <0.05% of all bladder tumors. PGL and UBPGL can be associated with SDHB, SDHD, NF1, and VHL gene variants, with the most common germline alterations found in SDHB and VHL. Case report We report a case of a 42-year-old woman who presented with menorrhagia/hematuria, uterine leiomyomas, as well as cardiac and bladder masses. The cardiac mass was favored to be a myxoma based on clinical findings, while the bladder mass was diagnosed as UBPGL. A novel SDHB mutation (c.642G>A, p Q214Q), detected in the UBPGL, was proven to be somatic. Although this variant was seemingly synonymous, it was predicted to have a loss of function due to the splice site effect, which was further supported by the immunohistochemical loss of SDHB. Conclusion This case highlights the challenges of diagnosing an extremely rare entity, bladder paraganglioma, with an emphasis on the multidisciplinary approach to navigate various clinical and imaging findings that may initially be misleading. In addition, a novel loss of function SDHB variant that could have been overlooked as a synonymous variant is herein reported, while also illustrating the importance of both germline and somatic mutation testing.
Collapse
Affiliation(s)
- Thao Nguyen
- Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Zehra Ordulu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sunaina Shrestha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Urja Patel
- Division of Endocrinology, University of Florida, Gainesville, FL, United States
| | - Paul L. Crispen
- Department of Urology, University of Florida, Gainesville, FL, United States
| | - Lisa Brown
- Department of Genetics, University of Florida, Gainesville, FL, United States
| | - Sara M. Falzarano
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Urology, University of Florida, Gainesville, FL, United States
- Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - Hans K. Ghayee
- Division of Endocrinology, University of Florida, Gainesville, FL, United States
- Division of Endocrinology, Malcom Randall Veterans Affairs (VA) Medical Center, Gainesville, FL, United States
| | | |
Collapse
|
10
|
Iguchi DYV, Martins Filho SN, Soares IC, Siqueira SAC, Alves VAF, Assato AK, Yang JH, Almeida MQ, Villares Fragoso MCB, Fagundes GFC, Mendonca BB, Lourenço Junior DM, Hoff AO, Castroneves LA, Ferraz-de-Souza B, Giannella MLCC, Pereira MAA. Identification of Predictors of Metastatic Potential in Paragangliomas to Develop a Prognostic Score (PSPGL). J Endocr Soc 2024; 8:bvae093. [PMID: 38799767 PMCID: PMC11112433 DOI: 10.1210/jendso/bvae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Indexed: 05/29/2024] Open
Abstract
Context Paragangliomas (PGLs) are rare tumors in adrenal and extra-adrenal locations. Metastasis are found in approximately 5% to 35% of PGLs, and there are no reliable predictors of metastatic disease. Objective This work aimed to develop a prognostic score of metastatic potential in PGLs. Methods A retrospective analysis was conducted of clinical data from a cohort with PGLs and tumor histological assessment. Patients were divided into metastatic PGL (presence of metastasis) and nonmetastatic PGL (absence of metastasis ≥96 months of follow-up) groups. Univariate and multivariable analysis were performed to identify predictors of metastatic potential. A prognostic score was developed based on coefficients of multivariable analysis. Kaplan-Meier curves were generated to estimate disease-specific survival (DSS). Results Out of 263 patients, 35 patients had metastatic PGL and 110 patients had nonmetastatic PGL. In multivariable analysis, 4 features were independently related to metastatic disease and composed the Prognostic Score of Paragangliomas (PSPGL): presence of central or confluent necrosis (33 points), more than 3 mitosis/10 high-power field (HPF) (28 points), extension into adipose tissue (20 points), and extra-adrenal location (19 points). A PSPGL of 24 or greater showed similar sensitivity with higher specificity than the Pheochromocytoma of the Adrenal Gland Scaled Score (PASS) and Grading System for Adrenal Pheochromocytoma and Paraganglioma (GAPP). PSPGL less than or equal to 20 was associated with a risk of metastasis of approximately 10%, whereas a PSPGL of 40 or greater was associated with approximately 80%. The presence of metastasis and Ki-67 of 3% or greater were related to lower DSS. Conclusion The PSPGL, composed of 4 easy-to-assess parameters, demonstrated good performance in predicting metastatic potential and good ability in estimating metastasis risk.
Collapse
Affiliation(s)
- Daniela Yone Veiga Iguchi
- Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | | | - Iberê Cauduro Soares
- Divisão de Anatomia Patológica, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Sheila Aparecida Coelho Siqueira
- Divisão de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Venâncio Avancini Ferreira Alves
- Laboratório de Investigaçãoc Médica LIM/14, Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Aline Kawassaki Assato
- Laboratório de Investigaçãoc Médica LIM/14, Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ji Hoon Yang
- Clínica de Endocrinologia, Hospital do Servidor Público Municipal de São Paulo, São Paulo 01532-000, Brazil
| | - Madson Q Almeida
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Maria Candida Barisson Villares Fragoso
- Laboratório de Hormônios e Genética Molecular LIM/42, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Gustavo Freitas Cardoso Fagundes
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Berenice B Mendonca
- Laboratório de Hormônios e Genética Molecular LIM/42, Laboratório de Sequenciamento em Larga Escala (SELA), Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Delmar Muniz Lourenço Junior
- Laboratório de Endocrinologia Molecular e Celular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ana O Hoff
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Luciana Audi Castroneves
- Divisão de Endocrinologia e Metabologia, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Bruno Ferraz-de-Souza
- Laboratório de Endocrinologia Celular e Molecular LIM/25, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo 01246-903, Brazil
- School of Medicine, University of Notre Dame Australia, Fremantle WA 6160, Australia
| | - Maria Lucia Cardillo Correa Giannella
- Laboratório de Carboidratos e Radioimunoensaio LIM/18, Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Maria Adelaide Albergaria Pereira
- Divisão de Endocrinologia e Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| |
Collapse
|
11
|
Jimenez C, Habra MA, Campbell MT, Tamsen G, Cruz-Goldberg D, Long J, Bassett R, Dantzer R, Balderrama-Brondani V, Varghese J, Lu Y. Cabozantinib in patients with unresectable and progressive metastatic phaeochromocytoma or paraganglioma (the Natalie Trial): a single-arm, phase 2 trial. Lancet Oncol 2024; 25:658-667. [PMID: 38608693 DOI: 10.1016/s1470-2045(24)00133-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Metastatic phaeochromocytomas and paragangliomas (MPPGs) are orphan diseases. Up to 50% of MPPGs are associated with germline pathogenic variants of the SDHB gene. These tumours and many non-familial MPPGs exhibit a phenotype that is characterised by abnormal angiogenesis. We aimed to assess the activity and safety of cabozantinib, an antiangiogenic multi-tyrosine kinase inhibitor, in patients with MPPGs. METHODS The Natalie Trial is a single-arm, phase 2 clinical trial being conducted at The University of Texas MD Anderson Cancer Center (Houston, TX, USA). Patients aged 18 years or older with histologically confirmed, progressive, and unresectable MPPGs, with an Eastern Cooperative Oncology Group performance status of 0-2, were treated with oral cabozantinib 60 mg/day. The primary endpoint was the investigator-assessed overall response rate per the Response Evaluation Criteria in Solid Tumours version 1.1 criteria. All outcomes were assessed in all evaluable participants who received any amount of study treatment. The trial is registered with ClinicalTrials.gov (NCT02302833) and is active but not recruiting. FINDINGS From March 10, 2015, to May 11, 2021, 17 patients (13 male participants and four female participants) were enrolled. The median follow-up was 25 months (IQR 18-49). The overall response rate was 25·0% (95% CI 7·3-52·4; four of 16 patients). Seven grade 3 adverse events were reported in six patients, including single cases of hand-and-foot syndrome, hypertension, rectal fistula, QT prolongation, and asymptomatic hypomagnesaemia, and two cases of asymptomatic elevations of amylase and lipase. There were no grade 4 adverse events and no patient died on-study. INTERPRETATION Cabozantinib shows promising activity in patients with MPPGs. FUNDING Team NAT Foundation, Margaret Cazalot, and Clarence P Cazalot.
Collapse
Affiliation(s)
- Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Mouhammed Amir Habra
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gina Tamsen
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Damaris Cruz-Goldberg
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vania Balderrama-Brondani
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeena Varghese
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine and Molecular Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Im K, Choi YJ, Kim DH, Kim DS, Ban K, Ji W, Baek IJ, Choi CM, Lee JC, Rho JK. AXL receptor tyrosine kinase inhibition improves the anti-tumor effects of CD8 + T cells by inducing CD103 + dendritic cell-mediated T cell priming. Biochem Biophys Res Commun 2023; 680:7-14. [PMID: 37703603 DOI: 10.1016/j.bbrc.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/23/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
AXL is a member of TAM receptor family and has been highlighted as a potential target for cancer treatment. Accumulating evidence has uncovered the critical role of the AXL signaling pathway in tumor growth, metastasis, and resistance against anti-cancer drugs, as well as its association with cancer immune escape. However, the function of AXL as a manipulator of the immune system in the tumor microenvironment (TME) remains unclear. Therefore, in this study, we investigated the impact of AXL on immune cells in the TME of a syngeneic tumor model using AXL knockout (AXL-/-) mice. Compared to AXL wild-type (AXL+/+) mice, tumor growth was significantly suppressed in AXL-/- mice, and an induced population of tumor-infiltrated CD8+ T cells and CD103+ dendritic cells (DCs) was observed. The change of CD8+ T cells and CD103+ DCs was also confirmed in tumor-draining lymph nodes (TdLN). In addition, the clonal expansion of OVA-specific CD8+ T cells was dominant in AXL-/- mice. Finally, anti-PD-1 treatment evidenced synergistic anti-cancer effects in AXL-/- mice. Overall, our data indicate that AXL signaling may inhibit the clonal expansion of tumor-specific CD8+ T cells through the regulation of the migration of CD8+ T cells and DCs in TME. Thus, AXL may be a powerful molecular target to improve anti-cancer effects through single or combined therapy with immune checkpoint inhibitors (ICI).
Collapse
Affiliation(s)
- Kyungtaek Im
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Yun Jung Choi
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Dong Ha Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Da-Som Kim
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Kyosun Ban
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Wonjun Ji
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - In-Jeoung Baek
- Department of Cell and Genetic Engineering, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Chang-Min Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea
| | - Jin Kyung Rho
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, 05505, South Korea.
| |
Collapse
|
13
|
Li L, Guan L, Tang Y, Zou Y, Zhong J, Qiu L. Research in the genetics of pheochromocytoma and paraganglioma: a bibliometric analysis from 2002 to 2022. Clin Exp Med 2023; 23:3969-3980. [PMID: 37103653 DOI: 10.1007/s10238-023-01049-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/17/2023] [Indexed: 04/28/2023]
Abstract
Over the past two decades, there has been a significant growth in articles focusing on the genetics of pheochromocytoma and paraganglioma (PPGL). We used bibliometric methods to investigate the historical changes and trend in PPGL research. There was a total of 1263 articles published in English from 2002 to 2022 included in our study. The number of annual publications and citations in this field has been increasing in the past 20 years. Furthermore, most of the publications originated from the European countries and the United States. The co-occurrence analysis showed close cooperation between different countries, institutions, or authors. The dual-map discipline analysis revealed that majority articles focused on four disciplines: #2 (Medicine, Medical, Clinical), #4 (Molecular, Biology, Immunology), #5 (Health, Nursing, Medicine), and #8 (Molecular, Biology, Genetics). The hotspot analysis revealed the keywords that have been landmark for PPGL genetics research in different time periods, and there was continued interest in gene mutations, especially on SDHX family genes. In conclusion, this study displays the current status of research and future trends in the genetics of PPGL. In future, more in-depth research should concentrate on crucial mutation genes and their specific mechanisms to assist in molecular target therapy. It is hoped that this study may help to provide directions for future research on genes and PPGL.
Collapse
Affiliation(s)
- Lei Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Lihua Guan
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Yueming Tang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Jian Zhong
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China.
| |
Collapse
|
14
|
Salomoni P, Flanagan AM, Cottone L. (B)On(e)-cohistones and the epigenetic alterations at the root of bone cancer. Cell Death Differ 2023:10.1038/s41418-023-01227-9. [PMID: 37828086 DOI: 10.1038/s41418-023-01227-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Identification of mutations in histones in a number of human neoplasms and developmental syndromes represents the most compelling evidence to date for a causal role of epigenetic perturbations in human disease. In most cases, these mutations have gain of function properties that cause deviation from normal developmental processes leading to embryo defects and/or neoplastic transformation. These exciting discoveries represent a step-change in our understanding of the role of chromatin (dys)regulation in development and disease. However, the mechanisms of action of oncogenic histone mutations (oncohistones) remain only partially understood. Here, we critically assess existing literature on oncohistones focussing mainly on bone neoplasms. We show how it is possible to draw parallels with some of the cell-autonomous mechanisms of action described in paediatric brain cancer, although the functions of oncohistones in bone tumours remain under-investigated. In this respect, it is becoming clear that histone mutations targeting the same residues display, at least in part, tissue-specific oncogenic mechanisms. Furthermore, it is emerging that cancer cells carrying oncohistones can modify the surrounding microenvironment to support growth and/or alter differentiation trajectories. A better understanding of oncohistone function in different neoplasms provide potential for identification of signalling that could be targeted therapeutically. Finally, we discuss some of the main concepts and future directions in this research area, while also drawing possible connections and parallels with other cancer epigenetic mechanisms.
Collapse
Affiliation(s)
- Paolo Salomoni
- Nuclear Function Group, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.
| | - Adrienne M Flanagan
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK
- Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Lucia Cottone
- Department of Pathology, UCL Cancer Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
15
|
Guo Q, Cheng ZM, Gonzalez-Cantú H, Rotondi M, Huelgas-Morales G, Ethiraj P, Qiu Z, Lefkowitz J, Song W, Landry BN, Lopez H, Estrada-Zuniga CM, Goyal S, Khan MA, Walker TJ, Wang E, Li F, Ding Y, Mulligan LM, Aguiar RCT, Dahia PLM. TMEM127 suppresses tumor development by promoting RET ubiquitination, positioning, and degradation. Cell Rep 2023; 42:113070. [PMID: 37659079 PMCID: PMC10637630 DOI: 10.1016/j.celrep.2023.113070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/06/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
The TMEM127 gene encodes a transmembrane protein of poorly known function that is mutated in pheochromocytomas, neural crest-derived tumors of adrenomedullary cells. Here, we report that, at single-nucleus resolution, TMEM127-mutant tumors share precursor cells and transcription regulatory elements with pheochromocytomas carrying mutations of the tyrosine kinase receptor RET. Additionally, TMEM127-mutant pheochromocytomas, human cells, and mouse knockout models of TMEM127 accumulate RET and increase its signaling. TMEM127 contributes to RET cellular positioning, trafficking, and lysosome-mediated degradation. Mechanistically, TMEM127 binds to RET and recruits the NEDD4 E3 ubiquitin ligase for RET ubiquitination and degradation via TMEM127 C-terminal PxxY motifs. Lastly, increased cell proliferation and tumor burden after TMEM127 loss can be reversed by selective RET inhibitors in vitro and in vivo. Our results define TMEM127 as a component of the ubiquitin system and identify aberrant RET stabilization as a likely mechanism through which TMEM127 loss-of-function mutations cause pheochromocytoma.
Collapse
Affiliation(s)
- Qianjin Guo
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Zi-Ming Cheng
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Hector Gonzalez-Cantú
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Matthew Rotondi
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Gabriela Huelgas-Morales
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Purushoth Ethiraj
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Zhijun Qiu
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Jonathan Lefkowitz
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Wan Song
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Bethany N Landry
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Hector Lopez
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Cynthia M Estrada-Zuniga
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Shivi Goyal
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Mohammad Aasif Khan
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA
| | - Timothy J Walker
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Exing Wang
- Department Cell Structure and Anatomy, UTHSCSA, San Antonio, TX, USA
| | - Faqian Li
- Department of Pathology, UTHSCSA, San Antonio, TX, USA
| | - Yanli Ding
- Department of Pathology, UTHSCSA, San Antonio, TX, USA
| | - Lois M Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Ricardo C T Aguiar
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA; Mays Cancer Center, UTHSCSA, San Antonio, TX, USA; South Texas Veterans Health Care System, Audie Murphy VA Hospital, San Antonio, TX 78229, USA
| | - Patricia L M Dahia
- Division of Hematology/Medical Oncology, Department of Medicine, University of Texas Health San Science Center at Antonio (UTHSCSA), San Antonio, TX, USA; Mays Cancer Center, UTHSCSA, San Antonio, TX, USA.
| |
Collapse
|
16
|
L Bello M, Mendes GEM, Silva ACR, Faria RX. Virtual screening indicates potential inhibitors of the P2X7 receptor. Comput Biol Med 2023; 164:107299. [PMID: 37552915 DOI: 10.1016/j.compbiomed.2023.107299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023]
Abstract
Anti-inflammatory agents can be synthetic or natural compounds and are often used to attenuate different levels of inflammation. Inflammatory diseases, due to the involvement of multiple systems, are becoming difficult to treat, involve long durations of therapy where applicable, have a high cost of management and have a deleterious impact on public health. The search for natural and synthetic compounds with anti-inflammatory activity is an important strategy in drug design. Bioactive synthetic drugs may be repurposed for other pharmacological applications, and natural product chemical structures offer unlimited opportunities for new drug discoveries due to the unparalleled availability of chemical diversity. Virtual screening of 2774 molecules on the mouse P2X7 protein showed that potential ligands are composed of five flavonoids (narirutin, diosmin, complanatuside, hesperidin, and oroxin B) and other drugs such as velpatasvir, itacitinib and lifitegrast. In vitro studies in mouse cells confirmed the inhibitory activity of the indicated ligands on the P2X7 receptor by applying virtual screening. The behavior of protein bonded to the ligands was verified by analysis of the molecular dynamic simulation trajectories for four of the most potent inhibitor compounds, indicating that the ligands velpatasvir, itacitinib, lithospermic acid and narirutin remained in the binding site indicated by molecular docking.
Collapse
Affiliation(s)
- Murilo L Bello
- Pharmaceutical Planning and Computer Simulation Laboratory, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme Eduardo M Mendes
- Pharmaceutical Planning and Computer Simulation Laboratory, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Postgraduate Program in Sciences and Biotechnology, Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Ana Cláudia R Silva
- Laboratory for Environmental Health Assessment and Promotion, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Robson X Faria
- Laboratory for Environmental Health Assessment and Promotion, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
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
|
18
|
Mellid S, Gil E, Letón R, Caleiras E, Honrado E, Richter S, Palacios N, Lahera M, Galofré JC, López-Fernández A, Calatayud M, Herrera-Martínez AD, Galvez MA, Matias-Guiu X, Balbín M, Korpershoek E, Lim ES, Maletta F, Lider S, Fliedner SMJ, Bechmann N, Eisenhofer G, Canu L, Rapizzi E, Bancos I, Robledo M, Cascón A. Co-occurrence of mutations in NF1 and other susceptibility genes in pheochromocytoma and paraganglioma. Front Endocrinol (Lausanne) 2023; 13:1070074. [PMID: 36760809 PMCID: PMC9905101 DOI: 10.3389/fendo.2022.1070074] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction The percentage of patients diagnosed with pheochromocytoma and paraganglioma (altogether PPGL) carrying known germline mutations in one of the over fifteen susceptibility genes identified to date has dramatically increased during the last two decades, accounting for up to 35-40% of PPGL patients. Moreover, the application of NGS to the diagnosis of PPGL detects unexpected co-occurrences of pathogenic allelic variants in different susceptibility genes. Methods Herein we uncover several cases with dual mutations in NF1 and other PPGL genes by targeted sequencing. We studied the molecular characteristics of the tumours with co-occurrent mutations, using omic tools to gain insight into the role of these events in tumour development. Results Amongst 23 patients carrying germline NF1 mutations, targeted sequencing revealed additional pathogenic germline variants in DLST (n=1) and MDH2 (n=2), and two somatic mutations in H3-3A and PRKAR1A. Three additional patients, with somatic mutations in NF1 were found carrying germline pathogenic mutations in SDHB or DLST, and a somatic truncating mutation in ATRX. Two of the cases with dual germline mutations showed multiple pheochromocytomas or extra-adrenal paragangliomas - an extremely rare clinical finding in NF1 patients. Transcriptional and methylation profiling and metabolite assessment showed an "intermediate signature" to suggest that both variants had a pathological role in tumour development. Discussion In conclusion, mutations affecting genes involved in different pathways (pseudohypoxic and receptor tyrosine kinase signalling) co-occurring in the same patient could provide a selective advantage for the development of PPGL, and explain the variable expressivity and incomplete penetrance observed in some patients.
Collapse
Affiliation(s)
- Sara Mellid
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Gil
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rocío Letón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Susan Richter
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nuria Palacios
- Endocrinology Department, University Hospital Puerta de Hierro, Madrid, Spain
| | - Marcos Lahera
- Endocrinology and Nutrition Department, La Princesa University Hospital, Madrid, Spain
| | - Juan C. Galofré
- Department of Endocrinology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Adriá López-Fernández
- Hereditary Cancer Genetics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Calatayud
- Department of Endocrinology and Nutrition, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - María A. Galvez
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Bellvitge University Hospital, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Milagros Balbín
- Molecular Oncology Laboratory, Instituto Universitario de Oncologia del Principado de Asturias, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Esther Korpershoek
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Eugénie S. Lim
- Department of Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Francesca Maletta
- Pathology Unit , Department of Laboratory Medicine, Azienda Ospedaliero-Universitaria (AOU) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Sofia Lider
- Endocrinology Department, National Institute of Endocrinology, Bucharest, Romania
| | | | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Letizia Canu
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Irina Bancos
- Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, United States
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Tabebi M, Söderkvist P, Gimm O. Nuclear and mitochondrial DNA alterations in pheochromocytomas and paragangliomas, and their potential treatment. Endocr Relat Cancer 2023; 30:ERC-22-0217. [PMID: 36219865 DOI: 10.1530/erc-22-0217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Mitochondrial DNA (mtDNA) alterations have been reported in different types of cancers and are suggested to play important roles in cancer development and metastasis. However, there is little information about its involvement in pheochromocytomas and paragangliomas (PCCs/PGLs) formation. PCCs and PGLs are rare endocrine tumors of the chromaffin cells in the adrenal medulla and extra-adrenal paraganglia that can synthesize and secrete catecholamines. Over the last 3 decades, the genetic background of about 60% of PCCs/PGLs involving nuclear DNA alterations has been determined. Recently, a study showed that mitochondrial alterations can be found in around 17% of the remaining PCCs/PGLs. In this review, we summarize recent knowledge regarding both nuclear and mitochondrial alterations and their involvement in PCCs/PGLs. We also provide brief insights into the genetics and the molecular pathways associated with PCCs/PGLs and potential therapeutical targets.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Peter Söderkvist
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, Linköping University, Linköping, Sweden
| | - Oliver Gimm
- Department of Surgery, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| |
Collapse
|
20
|
Martinelli S, Amore F, Canu L, Maggi M, Rapizzi E. Tumour microenvironment in pheochromocytoma and paraganglioma. Front Endocrinol (Lausanne) 2023; 14:1137456. [PMID: 37033265 PMCID: PMC10073672 DOI: 10.3389/fendo.2023.1137456] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Pheochromocytomas and Paragangliomas (Pheo/PGL) are rare catecholamine-producing tumours derived from adrenal medulla or from the extra-adrenal paraganglia respectively. Around 10-15% of Pheo/PGL develop metastatic forms and have a poor prognosis with a 37% of mortality rate at 5 years. These tumours have a strong genetic determinism, and the presence of succinate dehydrogenase B (SDHB) mutations are highly associated with metastatic forms. To date, no effective treatment is present for metastatic forms. In addition to cancer cells, the tumour microenvironment (TME) is also composed of non-neoplastic cells and non-cellular components, which are essential for tumour initiation and progression in multiple cancers, including Pheo/PGL. This review, for the first time, provides an overview of the roles of TME cells such as cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) on Pheo/PGL growth and progression. Moreover, the functions of the non-cellular components of the TME, among which the most representatives are growth factors, extracellular vesicles and extracellular matrix (ECM) are explored. The importance of succinate as an oncometabolite is emerging and since Pheo/PGL SDH mutated accumulate high levels of succinate, the role of succinate and of its receptor (SUCNR1) in the modulation of the carcinogenesis process is also analysed. Further understanding of the mechanism behind the complicated effects of TME on Pheo/PGL growth and spread could suggest novel therapeutic targets for further clinical treatments.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Francesca Amore
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Elena Rapizzi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- *Correspondence: Elena Rapizzi,
| |
Collapse
|
21
|
Advances in Adrenal and Extra-adrenal Paraganglioma: Practical Synopsis for Pathologists. Adv Anat Pathol 2023; 30:47-57. [PMID: 36136370 DOI: 10.1097/pap.0000000000000365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adrenal paraganglioma (or "pheochromocytoma") and extra-adrenal paraganglioma, collectively abbreviated PPGL, are rare but spectacular nonepithelial neuroendocrine neoplasms. These are the most inheritable neoplasia of all, with a metastatic potential in a varying degree. As of such, these lesions demand careful histologic, immunohistochemical, and genetic characterization to provide the clinical team with a detailed report taking into account the anticipated prognosis and risk of syndromic/inherited disease. While no histologic algorithm, immunohistochemical biomarker, or molecular aberration single-handedly can identify potentially lethal cases upfront, the combined analysis of various risk parameters may stratify PPGL patients more stringently than previously. Moreover, the novel 2022 WHO Classification of Endocrine and Neuroendocrine Tumors also brings some new concepts into play, not least the reclassification of special neuroendocrine neoplasms (cauda equina neuroendocrine tumor and composite gangliocytoma/neuroma-neuroendocrine tumor) previously thought to belong to the spectrum of PPGL. This review focuses on updated key diagnostic and prognostic concepts that will aid when facing this rather enigmatic tumor entity in clinical practice.
Collapse
|
22
|
Winzeler B, Tufton N, S. Lim E, Challis BG, Park S, Izatt L, Carroll PV, Velusamy A, Hulse T, Whitelaw BC, Martin E, Rodger F, Maranian M, Clark GR, A. Akker S, Maher ER, Casey RT. Investigating the role of somatic sequencing platforms for phaeochromocytoma and paraganglioma in a large UK cohort. Clin Endocrinol (Oxf) 2022; 97:448-459. [PMID: 34870338 PMCID: PMC9543043 DOI: 10.1111/cen.14639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 10/21/2021] [Indexed: 12/03/2022]
Abstract
OBJECTIVES Phaeochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumours with malignant potential and a hereditary basis in almost 40% of patients. Germline genetic testing has transformed the management of PPGL enabling stratification of surveillance approaches, earlier diagnosis and predictive testing of at-risk family members. Recent studies have identified somatic mutations in a further subset of patients, indicating that molecular drivers at either a germline or tumour level can be identified in up to 80% of PPGL cases. The aim of this study was to investigate the clinical utility of somatic sequencing in a large cohort of patients with PPGL in the United Kingdom. DESIGN AND PATIENTS Prospectively collected matched germline and tumour samples (development cohort) and retrospectively collected tumour samples (validation cohort) of patients with PPGL were investigated. MEASUREMENTS Clinical characteristics of patients were assessed and tumour and germline DNA was analysed using a next-generation sequencing strategy. A screen for variants within 'mutation hotspots' in 68 human cancer genes was performed. RESULTS Of 141 included patients, 45 (32%) had a germline mutation. In 37 (26%) patients one or more driver somatic variants were identified including 26 likely pathogenic or pathogenic variants and 19 variants of uncertain significance. Pathogenic somatic variants, observed in 25 (18%) patients, were most commonly identified in the VHL, NF1, HRAS and RET genes. Pathogenic somatic variants were almost exclusively identified in patients without a germline mutation (all but one), suggesting that somatic sequencing is likely to be most informative for those patients with negative germline genetic test results. CONCLUSIONS Somatic sequencing may further stratify surveillance approaches for patients without a germline genetic driver and may also inform targeted therapeutic strategies for patients with metastatic disease.
Collapse
Affiliation(s)
- Bettina Winzeler
- Department of Endocrinology, Diabetology and MetabolismUniversity Hospital BaselBaselSwitzerland
- Department of Clinical ResearchUniversity of BaselBaselSwitzerland
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Nicola Tufton
- Department of EndocrinologySt. Bartholomew's Hospital, Barts Health NHS TrustLondonUK
- Department of Endocrinology, William Harvey Research InstituteQueen Mary University of LondonLondon
| | - Eugenie S. Lim
- Department of EndocrinologySt. Bartholomew's Hospital, Barts Health NHS TrustLondonUK
- Department of Endocrinology, William Harvey Research InstituteQueen Mary University of LondonLondon
| | - Ben G. Challis
- Department of Endocrinology, Cambridge University HospitalNHS Foundation TrustCambridgeUK
| | - Soo‐Mi Park
- Department of Clinical Genetics, Cambridge University HospitalNHS Foundation TrustCambridgeUK
| | - Louise Izatt
- Department of Clinical GeneticsGuy's and St. Thomas' NHS Foundation TrustLondonUK
| | - Paul V. Carroll
- Department of EndocrinologyGuy's and St. Thomas' NHS Foundation TrustLondonUK
| | - Anand Velusamy
- Department of EndocrinologyGuy's and St. Thomas' NHS Foundation TrustLondonUK
| | - Tony Hulse
- Department of Paediatric EndocrinologyEvelina London Children's Hospital, Guy's and St. Thomas' NHS Foundation TrustLondonUK
| | | | - Ezequiel Martin
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
- Oncology Department, Cancer Molecular Diagnostics LaboratoryUniversity of CambridgeCambridgeUK
| | - Fay Rodger
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Melanie Maranian
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Graeme R. Clark
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Scott A. Akker
- Department of EndocrinologySt. Bartholomew's Hospital, Barts Health NHS TrustLondonUK
- Department of Endocrinology, William Harvey Research InstituteQueen Mary University of LondonLondon
| | - Eamonn R. Maher
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
| | - Ruth T. Casey
- Department of Medical Genetics, and Cancer Research, UK Cambridge Centre, University of CambridgeCambridge Biomedical CampusCambridgeUK
- Department of Endocrinology, Cambridge University HospitalNHS Foundation TrustCambridgeUK
| |
Collapse
|
23
|
Ma X, Cui Y, Gao Y, Zhang X, Nie M, Tong A. Fumarate hydratase gene germline variants and mosaicism associated with pheochromocytoma and paraganglioma. Ann N Y Acad Sci 2022; 1516:262-270. [PMID: 35821608 DOI: 10.1111/nyas.14866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fumarate hydratase (FH) catalyzes the conversion of fumaric acid to L-malic acid. Heterozygous variants of the human fumarate hydratase gene (FH) predispose to hereditary leiomyomatosis and renal cell cancer and, rarely, pheochromocytoma/paraganglioma (PPGL). No mosaic variant in FH has been reported yet. Using next-generation sequencing, five individuals with FH variants were found in 319 PPGL patients. Immunohistochemistry staining and loss of heterozygosity analysis in tumor tissues were performed to determine the pathogenicity of the variants. Deep targeted sequencing was performed on the peripheral blood DNA of a pheochromocytoma (PCC) patient with uterine leiomyomas. Finally, two of the five variants were found to be pathogenic. A germline variant (c.817G>A, p.Ala273Thr) was found in a patient with a PPGL family history. A mosaic variant (c.206G>A, p.Gly69Asp) with an allelic ratio of 5% in blood DNA was confirmed in the PCC patient with uterine leiomyomas. No metastatic PPGL was observed in the two PPGL patients with FH pathogenic variants. In summary, we report mosaicism in FH and the first PPGL pedigree with an FH pathogenic germline variant. Both germline variants and mosaicism should be taken into account during genetic testing.
Collapse
Affiliation(s)
- Xiaosen Ma
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunying Cui
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yinjie Gao
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuebin Zhang
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Nie
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Anli Tong
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Zhang J, Li M, Pang Y, Wang C, Wu J, Cheng Z, Li X, Lu Z, Liu Y, Guo J, Chen X, He Y, Guan X, Xu X, Wang Y, Liu J, Guo W, Hou Y, Liu L, Jiang J, Gao X. Genetic Characteristics of Incidental Pheochromocytoma and Paraganglioma. J Clin Endocrinol Metab 2022; 107:e1835-e1842. [PMID: 35106577 DOI: 10.1210/clinem/dgac058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Pheochromocytomas and paragangliomas (PPGLs) are being increasingly discovered by imaging performed for unrelated conditions. The genetic landscape of incidental PPGLs remains to be elucidated. OBJECTIVE We aimed to describe the genetic characteristics of PPGLs discovered incidentally in a large PPGL cohort. METHODS This retrospective cross-sectional study included 697 patients with pathology confirmed PPGLs, including 283 incidentalomas and 414 nonincidentalomas, at 2 tertiary care centers in China in 2009-2019. Tumor DNA samples were sequenced by next-generation sequencing. Identified genetic mutations were confirmed by Sanger sequencing and tested in 277 available matched blood DNA samples. RESULTS There was a lower proportion of patients with mutations identified (53% vs 63.3%; P = 0.0067) in incidental than nonincidental PPGLs. In incidental PPGLs, HRAS (11.7%), FGFR1 (11%), and RET (9.2%) were the top 3 mutated genes, whereas HRAS (17.9%), VHL (9.2%), and NF-1 (8.7%) exhibited the highest rate of mutations in nonincidental PPGLs. In incidental pheochromocytomas, the most frequently mutated genes were RET (10.9%), HRAS (10.4%), and VHL (8.6%), while in incidental paragangliomas, FGFR1 (32.8%), HRAS (16.4%), and EPAS1 (9.8%) topped the list. The frequency of NF-1 mutations was significantly lower in incidental than nonincidental pheochromocytomas (4.1% vs 11%; P = 0.0042), while FGFR1 mutations were far more common in incidental than nonincidental paragangliomas (32.8% vs 15.3%; P = 0.0076). CONCLUSION More than half of patients with incidental PPGLs had mutations in common susceptibility genes. The search for susceptibility genes should take both the mode of discovery (incidental vs nonincidental) and tumor location (pheochromocytoma vs paraganglioma) into consideration.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Minghao Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Cikui Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jingjing Wu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Ziyun Cheng
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Zhiqiang Lu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiao Guan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaowen Xu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yong Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiahao Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 361015, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
25
|
Nölting S, Bechmann N, Taieb D, Beuschlein F, Fassnacht M, Kroiss M, Eisenhofer G, Grossman A, Pacak K. Personalized Management of Pheochromocytoma and Paraganglioma. Endocr Rev 2022; 43:199-239. [PMID: 34147030 PMCID: PMC8905338 DOI: 10.1210/endrev/bnab019] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Pheochromocytomas/paragangliomas are characterized by a unique molecular landscape that allows their assignment to clusters based on underlying genetic alterations. With around 30% to 35% of Caucasian patients (a lower percentage in the Chinese population) showing germline mutations in susceptibility genes, pheochromocytomas/paragangliomas have the highest rate of heritability among all tumors. A further 35% to 40% of Caucasian patients (a higher percentage in the Chinese population) are affected by somatic driver mutations. Thus, around 70% of all patients with pheochromocytoma/paraganglioma can be assigned to 1 of 3 main molecular clusters with different phenotypes and clinical behavior. Krebs cycle/VHL/EPAS1-related cluster 1 tumors tend to a noradrenergic biochemical phenotype and require very close follow-up due to the risk of metastasis and recurrence. In contrast, kinase signaling-related cluster 2 tumors are characterized by an adrenergic phenotype and episodic symptoms, with generally a less aggressive course. The clinical correlates of patients with Wnt signaling-related cluster 3 tumors are currently poorly described, but aggressive behavior seems likely. In this review, we explore and explain why cluster-specific (personalized) management of pheochromocytoma/paraganglioma is essential to ascertain clinical behavior and prognosis, guide individual diagnostic procedures (biochemical interpretation, choice of the most sensitive imaging modalities), and provide personalized management and follow-up. Although cluster-specific therapy of inoperable/metastatic disease has not yet entered routine clinical practice, we suggest that informed personalized genetic-driven treatment should be implemented as a logical next step. This review amalgamates published guidelines and expert views within each cluster for a coherent individualized patient management plan.
Collapse
Affiliation(s)
- Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), CH-8091 Zurich, Switzerland.,Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, 13273 Marseille, France
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), CH-8091 Zurich, Switzerland.,Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany.,Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX2 6HG, UK.,Centre for Endocrinology, Barts and the London School of Medicine, London EC1M 6BQ, UK.,ENETS Centre of Excellence, Royal Free Hospital, London NW3 2QG, UK
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD 20847, USA
| |
Collapse
|
26
|
Genetics of Pheochromocytomas and Paragangliomas Determine the Therapeutical Approach. Int J Mol Sci 2022; 23:ijms23031450. [PMID: 35163370 PMCID: PMC8836037 DOI: 10.3390/ijms23031450] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Pheochromocytomas and paragangliomas are the most heritable endocrine tumors. In addition to the inherited mutation other driver mutations have also been identified in tumor tissues. All these genetic alterations are clustered in distinct groups which determine the pathomechanisms. Most of these tumors are benign and their surgical removal will resolve patient management. However, 5–15% of them are malignant and therapeutical possibilities for them are limited. This review provides a brief insight about the tumorigenesis associated with pheochromocytomas/paragangliomas in order to present them as potential therapeutical targets.
Collapse
|
27
|
Jhawar S, Arakawa Y, Kumar S, Varghese D, Kim YS, Roper N, Elloumi F, Pommier Y, Pacak K, Del Rivero J. New Insights on the Genetics of Pheochromocytoma and Paraganglioma and Its Clinical Implications. Cancers (Basel) 2022; 14:cancers14030594. [PMID: 35158861 PMCID: PMC8833412 DOI: 10.3390/cancers14030594] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pheochromocytoma and paraganglioma (together PPGL) are rare neuroendocrine tumors that arise from chromaffin tissue and produce catecholamines. Approximately 40% of cases of PPGL carry a germline mutation, suggesting that they have a high degree of heritability. The underlying mutation influences the PPGL clinical presentation such as cell differentiation, specific catecholamine production, tumor location, malignant potential and genetic anticipation, which helps to better understand the clinical course and tailor treatment accordingly. Genetic testing for pheochromocytoma and paraganglioma allows an early detection of hereditary syndromes and facilitates a close follow-up of high-risk patients. In this review article, we present the most recent advances in the field of genetics and we discuss the latest guidelines on the surveillance of asymptomatic SDHx mutation carriers. Abstract Pheochromocytomas (PHEOs) and paragangliomas (PGLs) are rare neuroendocrine tumors that arise from chromaffin cells. PHEOs arise from the adrenal medulla, whereas PGLs arise from the neural crest localized outside the adrenal gland. Approximately 40% of all cases of PPGLs (pheochromocytomas/paragangliomas) are associated with germline mutations and 30–40% display somatic driver mutations. The mutations associated with PPGLs can be classified into three groups. The pseudohypoxic group or cluster I includes the following genes: SDHA, SDHB, SDHC, SDHD, SDHAF2, FH, VHL, IDH1/2, MHD2, EGLN1/2 and HIF2/EPAS; the kinase group or cluster II includes RET, NF1, TMEM127, MAX and HRAS; and the Wnt signaling group or cluster III includes CSDE1 and MAML3. Underlying mutations can help understand the clinical presentation, overall prognosis and surveillance follow-up. Here we are discussing the new genetic insights of PPGLs.
Collapse
Affiliation(s)
- Sakshi Jhawar
- Life Bridge Health Center, Internal Medicine Program, Sinai Hospital of Baltimore, Baltimore, MD 21215, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Suresh Kumar
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Diana Varghese
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yoo Sun Kim
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nitin Roper
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Fathi Elloumi
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Wągrodzki M, Tysarowski A, Seliga K, Wojnowska A, Stepaniuk M, Castañeda Wysocka P, Makuła D, Pieńkowski A, Szostakowski B, Zub R, Rutkowski P. Diagnostic Utility of Genetic and Immunohistochemical H3-3A Mutation Analysis in Giant Cell Tumour of Bone. Int J Mol Sci 2022; 23:ijms23020969. [PMID: 35055156 PMCID: PMC8778699 DOI: 10.3390/ijms23020969] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
To validate the reliability and implementation of an objective diagnostic method for giant cell tumour of bone (GCTB). H3-3A gene mutation testing was performed using two different methods, Sanger sequencing and immunohistochemical (IHC) assays. A total of 214 patients, including 120 with GCTB and 94 with other giant cell-rich bone lesions, participated in the study. Sanger sequencing and IHC with anti-histone H3.3 G34W and G34V antibodies were performed on formalin-fixed, paraffin-embedded tissues, which were previously decalcified in EDTA if needed. The sensitivity and specificity of the molecular method was 100% (95% CI: 96.97–100%) and 100% (95% CI: 96.15–100%), respectively. The sensitivity and specificity of IHC was 94.32% (95% CI: 87.24–98.13%) and 100% (95% CI: 93.94–100.0%), respectively. P.G35 mutations were discovered in 2/9 (22.2%) secondary malignant GCTBs and 9/13 (69.2%) GCTB after denosumab treatment. We confirmed in a large series of patients that evaluation of H3-3A mutational status using direct sequencing is a reliable tool for diagnosing GCTB, and it should be incorporated into the diagnostic algorithm. Additionally, we discovered IHC can be used as a screening tool. Proper tissue processing and decalcification are necessary. The presence of the H3-3A mutation did not exclude malignant GCTB. Denosumab did not eradicate the neoplastic cell population of GCTB.
Collapse
Affiliation(s)
- Michał Wągrodzki
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence: ; Tel.: +48-537484741
| | - Andrzej Tysarowski
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (K.S.); (A.W.); (R.Z.)
| | - Katarzyna Seliga
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (K.S.); (A.W.); (R.Z.)
| | - Aneta Wojnowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (K.S.); (A.W.); (R.Z.)
| | - Maria Stepaniuk
- Department of Pathology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Patrycja Castañeda Wysocka
- Department of Radiology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (P.C.W.); (D.M.)
| | - Donata Makuła
- Department of Radiology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (P.C.W.); (D.M.)
| | - Andrzej Pieńkowski
- Department of Bone/Soft Tissue Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (B.S.); (P.R.)
| | - Bartłomiej Szostakowski
- Department of Bone/Soft Tissue Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (B.S.); (P.R.)
| | - Renata Zub
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (K.S.); (A.W.); (R.Z.)
| | - Piotr Rutkowski
- Department of Bone/Soft Tissue Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.P.); (B.S.); (P.R.)
| |
Collapse
|
29
|
Flores SK, Estrada-Zuniga CM, Thallapureddy K, Armaiz-Peña G, Dahia PLM. Insights into Mechanisms of Pheochromocytomas and Paragangliomas Driven by Known or New Genetic Drivers. Cancers (Basel) 2021; 13:cancers13184602. [PMID: 34572828 PMCID: PMC8467373 DOI: 10.3390/cancers13184602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/12/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Pheochromocytomas and paragangliomas are rare neuroendocrine tumors that are often hereditary. Although research has advanced considerably, significant gaps still persist in understanding risk factors, predicting metastatic potential and treating aggressive tumors. The study of rare mutations can provide new insights into how pheochromocytomas and paragangliomas develop. In this review, we provide examples of such rare events and how they can inform our understanding of the spectrum of mutations that can lead to these tumors and improve our ability to provide a genetic diagnosis. Abstract Pheochromocytomas and paragangliomas are rare tumors of neural crest origin. Their remarkable genetic diversity and high heritability have enabled discoveries of bona fide cancer driver genes with an impact on diagnosis and clinical management and have consistently shed light on new paradigms in cancer. In this review, we explore unique mechanisms of pheochromocytoma and paraganglioma initiation and management by drawing from recent examples involving rare mutations of hypoxia-related genes VHL, EPAS1 and SDHB, and of a poorly known susceptibility gene, TMEM127. These models expand our ability to predict variant pathogenicity, inform new functional domains, recognize environmental-gene connections, and highlight persistent therapeutic challenges for tumors with aggressive behavior.
Collapse
Affiliation(s)
- Shahida K. Flores
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (S.K.F.); (C.M.E.-Z.); (K.T.); (G.A.-P.)
| | - Cynthia M. Estrada-Zuniga
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (S.K.F.); (C.M.E.-Z.); (K.T.); (G.A.-P.)
| | - Keerthi Thallapureddy
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (S.K.F.); (C.M.E.-Z.); (K.T.); (G.A.-P.)
| | - Gustavo Armaiz-Peña
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (S.K.F.); (C.M.E.-Z.); (K.T.); (G.A.-P.)
| | - Patricia L. M. Dahia
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (S.K.F.); (C.M.E.-Z.); (K.T.); (G.A.-P.)
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Correspondence:
| |
Collapse
|
30
|
Pozdeyev N, Fishbein L, Gay LM, Sokol ES, Hartmaier R, Ross JS, Darabi S, Demeure MJ, Kar A, Foust L, Koc K, Bowles DW, Leong S, Wierman ME, Kiseljak-Vassiliades K. Targeted genomic analysis of 364 adrenocortical carcinomas. Endocr Relat Cancer 2021; 28:671-681. [PMID: 34410225 PMCID: PMC8384129 DOI: 10.1530/erc-21-0040] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022]
Abstract
Despite recent advances in elucidating molecular pathways underlying adrenocortical carcinoma (ACC), this orphan malignancy is associated with poor survival. Identification of targetable genomic alterations is critical to improve outcomes. The objective of this study was to characterize the genomic profile of a large cohort of patient ACC samples to identify actionable genomic alterations. Three hundred sixty-four individual patient ACC tumors were analyzed. The median age of the cohort was 52 years and 60.9% (n = 222) were female. ACC samples had common alterations in epigenetic pathways with 38% of tumors carrying alterations in genes involved in histone modification, 21% in telomere lengthening, and 21% in SWI/SNF complex. Tumor suppressor genes and WNT signaling pathway were each mutated in 51% of tumors. Fifty (13.7%) ACC tumors had a genomic alteration in genes involved in the DNA mismatch repair (MMR) pathway with many tumors also displaying an unusually high number of mutations and a corresponding MMR mutation signature. In addition, genomic alterations in several genes not previously associated with ACC were observed, including IL7R, LRP1B, FRS2 mutated in 6, 8 and 4% of tumors, respectively. In total, 58.5% of ACC (n = 213) had at least one potentially actionable genomic alteration in 46 different genes. As more than half of ACC have one or more potentially actionable genomic alterations, this highlights the value of targeted sequencing for this orphan cancer with a poor prognosis. In addition, significant incidence of MMR gene alterations suggests that immunotherapy is a promising therapeutic for a considerable subset of ACC patients.
Collapse
Affiliation(s)
- Nikita Pozdeyev
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
- Division of Biomedical Informatics & Personalized Medicine, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Lauren Fishbein
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
- Division of Biomedical Informatics & Personalized Medicine, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | | | | | | | - Jeffrey S. Ross
- Foundation Medicine Inc. Cambridge Massachusetts
- Departments of Pathology and Urology, Upstate Medical University, Syracuse, New York
| | - Sourat Darabi
- Hoag Family Center Institute, Newport Beach, California
| | - Michael J. Demeure
- Hoag Family Center Institute, Newport Beach, California
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Adwitiya Kar
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Lindsey Foust
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Katrina Koc
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Daniel W. Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Stephen Leong
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
| | - Margaret E. Wierman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
- Research Service Veterans Affairs Medical Center, Aurora Colorado 80045
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine at Colorado Anschutz Medical Campus Aurora, Colorado
- Research Service Veterans Affairs Medical Center, Aurora Colorado 80045
| |
Collapse
|
31
|
A Critical Appraisal of Contemporary and Novel Biomarkers in Pheochromocytomas and Adrenocortical Tumors. BIOLOGY 2021; 10:biology10070580. [PMID: 34201922 PMCID: PMC8301201 DOI: 10.3390/biology10070580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022]
Abstract
Simple Summary Pheochromocytomas/paragangliomas (PPGLs) and adrenocortical tumors are neoplasms that originate from different regions of the adrenal gland and display significant heterogeneity with respect to their biological and clinical behavior. They may be sporadic or develop in the context of hereditary syndromes. Adrenocortical tumors are mostly benign but less than 2% are carcinomas associated with a poor outcome when diagnosed in advanced disease. The majority of PPGLS are benign, but a subset may develop metastatic disease. In particular, for PPGLs, it is mandatory to identify biomarkers of high sensitivity and specificity that lead to accurate diagnosis and predict patients with a malignant potential that would benefit from aggressive surveillance and administration of early treatment. Current biomarkers include mostly histopathological and genetic parameters but none of them can be considered independent predictive factors. Recent genomic studies have implemented new molecular biomarkers of high accuracy for the diagnosis and management of PPGLs and adrenocortical tumors. In this review, we summarize the current and novel biomarkers that may be considered valuable tools for diagnosis and prediction of malignancy in patients with PPGLs and adrenocortical tumors. Abstract Pheochromocytomas/Paragangliomas (PPGLs) and adrenocortical tumors are rare neoplasms with significant heterogeneity in their biologic and clinical behavior. Current diagnostic and predictive biomarkers include hormone secretion, as well as histopathological and genetic features. PPGL diagnosis is based on biochemical measurement of catecholamines/metanephrines, while histopathological scoring systems have been proposed to predict the risk of malignancy. Adrenocortical tumors are mostly benign, but some can be malignant. Currently, the stage of disease at diagnosis and tumor grade, appear to be the most powerful prognostic factors. However, recent genomic and proteomic studies have identified new genetic and circulating biomarkers, including genes, immunohistochemical markers and micro-RNAs that display high specificity and sensitivity as diagnostic or prognostic tools. In addition, new molecular classifications have been proposed that divide adrenal tumors in distinct subgroups with different clinical outcomes.
Collapse
|
32
|
Leske H, Dalgleish R, Lazar AJ, Reifenberger G, Cree IA. A common classification framework for histone sequence alterations in tumours: an expert consensus proposal. J Pathol 2021; 254:109-120. [PMID: 33779999 DOI: 10.1002/path.5666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 12/17/2022]
Abstract
The description of genetic alterations in tumours is of increasing importance. In human genetics, and in pathology reports, sequence alterations are given using the human genome variation society (HGVS) guidelines for the description of such variants. However, there is less adherence to these guidelines for sequence variations in histone genes. Due to early cleavage of the N-terminal methionine in most histones, the description of histone sequence alterations follows their own nomenclature and differs from the HGVS-compliant numbering by omitting this first amino acid. Next generation sequencing reports, however, follow the HGVS guidelines and as a result, an unambiguous description of sequence variants in histones cannot be provided. The coexistence of these two nomenclatures leads to confusions for pathologists, oncologists, and researchers. This review provides an overview of tumour entities with sequence alterations of the H3-3A gene (HGNC ID = HGNC:4764), highlights the problems associated with the coexistence of these two nomenclatures, and proposes a standard for the reporting of histone sequence variants that allows an unambiguous description of these variants according to HGVS principles. We hope that scientific journals will adopt the new notation, and that both geneticists and pathologists will include it in their reports. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Henning Leske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- University of Oslo (UiO), Oslo, Norway
| | - Raymond Dalgleish
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Ian A Cree
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France
| |
Collapse
|
33
|
Abstract
Abdominal paragangliomas and pheochromocytomas (PPGLs) are rare neuroendocrine tumors of the infradiaphragmatic paraganglia and adrenal medulla, respectively. Although few pathologists outside of endocrine tertiary centers will ever diagnose such a lesion, the tumors are well known through the medical community-possible due to a combination of the sheer rarity, their often-spectacular presentation due to excess catecholamine secretion as well as their unrivaled coupling to constitutional susceptibility gene mutations and hereditary syndromes. All PPGLs are thought to harbor malignant potential, and therefore pose several challenges to the practicing pathologist. Specifically, a responsible diagnostician should recognize both the capacity and limitations of histological, immunohistochemical, and molecular algorithms to pinpoint high risk for future metastatic disease. This focused review aims to provide the surgical pathologist with a condensed update regarding the current strategies available in order to deliver an accurate prognostication of these enigmatic lesions.
Collapse
Affiliation(s)
- C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden.
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW This review summarizes our current understanding of germline and somatic genetics and genomics of pheochromocytomas and paragangliomas (PCC/PGL), describes existing knowledge gaps, and discusses future research directions. RECENT FINDINGS Germline pathogenic variants (PVs) are found in up to 40% of those with PCC/PGL. Tumors with germline PVs are broadly categorized as Cluster 1 (pseudohypoxia), including those with SDH, VHL, FH, and EPAS1 PVs, or Cluster 2 (kinase signaling) including those with NF1, RET, TMEM127, and MAX PVs. Somatic driver mutations exist in some of the same genes (RET, VHL, NF1, EPAS1) as well as in additional genes including HRAS, CSDE1 and genes involved in cell immortalization (ATRX and TERT). Other somatic driver events include recurrent fusion genes involving MAML3. SUMMARY PCC/PGL have the highest association with germline PVs of all human solid tumors. Expanding our understanding of the molecular pathogenesis of PCC/PGL is essential to advancements in diagnosis and surveillance and the development of novel therapies for these unique tumors.
Collapse
Affiliation(s)
- Heather Wachtel
- Hospital of the University of Pennsylvania, Department of Surgery, Division of Endocrine and Oncologic Surgery and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lauren Fishbein
- University of Colorado School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Diabetes and the Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
35
|
Mutation Profile of Aggressive Pheochromocytoma and Paraganglioma with Comparison of TCGA Data. Cancers (Basel) 2021; 13:cancers13102389. [PMID: 34069252 PMCID: PMC8156611 DOI: 10.3390/cancers13102389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Pheochromocytomas and paragangliomas (PPGLs) are neuroendocrine tumors arising from chromaffin cells of the adrenal medulla, or extra-adrenal paraganglia, respectively. In PPGLs, germline or somatic mutations in one of the known susceptibility genes are identified in up to 60% patients. Recent WHO classification defines that all PPGLs can have metastatic potential. The term, ‘malignant’ is replaced with ‘metastatic’ in this group of tumors. However, the peculiar genetic events that drive the aggressive behavior, including metastasis in PPGLs are yet poorly understood. We performed targeted next-generation sequencing analysis to characterize the mutation profile in fifteen aggressive PPGL patients and compared accessible data of aggressive PPGLs from The Cancer Genome Atlas (TCGA) with findings of our cohort. This targeted mutational analysis might expand the mutation profile of aggressive PPGLs, and may also be useful in detecting the possible experimental therapeutic options or predicting poor prognosis. Abstract In pheochromocytoma and paraganglioma (PPGL), germline or somatic mutations in one of the known susceptibility genes are identified in up to 60% patients. However, the peculiar genetic events that drive the aggressive behavior including metastasis in PPGL are poorly understood. We performed targeted next-generation sequencing analysis to characterize the mutation profile in fifteen aggressive PPGL patients and compared accessible data of aggressive PPGLs from The Cancer Genome Atlas (TCGA) with findings of our cohort. A total of 115 germline and 34 somatic variants were identified with a median 0.58 per megabase tumor mutation burden in our cohort. The most frequent mutation was SDHB germline mutation (27%) and the second frequent mutations were somatic mutations for SETD2, NF1, and HRAS (13%, respectively). Patients were subtyped into three categories based on the kind of mutated genes: pseudohypoxia (n = 5), kinase (n = 5), and unknown (n = 5) group. In copy number variation analysis, deletion of chromosome arm 1p harboring SDHB gene was the most frequently observed. In our cohort, SDHB mutation and pseudohypoxia subtype were significantly associated with poor overall survival. In conclusion, subtyping of mutation profile can be helpful in aggressive PPGL patients with heterogeneous prognosis to make relevant follow-up plan and achieve proper treatment.
Collapse
|
36
|
Garcia-Carbonero R, Matute Teresa F, Mercader-Cidoncha E, Mitjavila-Casanovas M, Robledo M, Tena I, Alvarez-Escola C, Arístegui M, Bella-Cueto MR, Ferrer-Albiach C, Hanzu FA. Multidisciplinary practice guidelines for the diagnosis, genetic counseling and treatment of pheochromocytomas and paragangliomas. Clin Transl Oncol 2021; 23:1995-2019. [PMID: 33959901 PMCID: PMC8390422 DOI: 10.1007/s12094-021-02622-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors that arise from chromaffin cells of the adrenal medulla and the sympathetic/parasympathetic neural ganglia, respectively. The heterogeneity in its etiology makes PPGL diagnosis and treatment very complex. The aim of this article was to provide practical clinical guidelines for the diagnosis and treatment of PPGLs from a multidisciplinary perspective, with the involvement of the Spanish Societies of Endocrinology and Nutrition (SEEN), Medical Oncology (SEOM), Medical Radiology (SERAM), Nuclear Medicine and Molecular Imaging (SEMNIM), Otorhinolaryngology (SEORL), Pathology (SEAP), Radiation Oncology (SEOR), Surgery (AEC) and the Spanish National Cancer Research Center (CNIO). We will review the following topics: epidemiology; anatomy, pathology and molecular pathways; clinical presentation; hereditary predisposition syndromes and genetic counseling and testing; diagnostic procedures, including biochemical testing and imaging studies; treatment including catecholamine blockade, surgery, radiotherapy and radiometabolic therapy, systemic therapy, local ablative therapy and supportive care. Finally, we will provide follow-up recommendations.
Collapse
Affiliation(s)
- R Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Avda Cordoba km 5.4, 28041, Madrid, Spain.
| | - F Matute Teresa
- Radiology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - E Mercader-Cidoncha
- Endocrine and Metabolic Surgery Unit, General and Digestive Surgery Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - M Mitjavila-Casanovas
- Nuclear Medicine Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain.,Grupo de Trabajo de Endocrino de la SEMNIM, Madrid, Spain
| | - M Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - I Tena
- Scientific Department, Medica Scientia Innovation Research (MedSIR CORP), Ridgewood, NJ, USA.,Medical Oncology Department, Hospital Provincial, Castellon, Spain
| | - C Alvarez-Escola
- Neuroendocrinology Unit, Endocrinology and Nutrition Department, Hospital Universitario la Paz, Madrid, Spain
| | - M Arístegui
- ENT Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - M R Bella-Cueto
- Pathology Department, Hospital Universitario Parc Taulí, Sabadell, Institut D'Investigació I Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Ferrer-Albiach
- Radiation Oncology Department, Hospital Provincial Castellón, Castellón, Spain
| | - F A Hanzu
- Endocrinology and Nutrition Department, Hospital Clinic Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| |
Collapse
|
37
|
Amatori S, Tavolaro S, Gambardella S, Fanelli M. The dark side of histones: genomic organization and role of oncohistones in cancer. Clin Epigenetics 2021; 13:71. [PMID: 33827674 PMCID: PMC8025322 DOI: 10.1186/s13148-021-01057-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Background The oncogenic role of histone mutations is one of the most relevant discovery in cancer epigenetics. Recurrent mutations targeting histone genes have been described in pediatric brain tumors, chondroblastoma, giant cell tumor of bone and other tumor types. The demonstration that mutant histones can be oncogenic and drive the tumorigenesis in pediatric tumors, led to the coining of the term “oncohistones.” The first identified histone mutations were localized at or near residues normally targeted by post-translational modifications (PTMs) in the histone N-terminal tails and suggested a possible interference with histone PTMs regulation and reading. Main body In this review, we describe the peculiar organization of the multiple genes that encode histone proteins, and the latter advances in both the identification and the biological role of histone mutations in cancer. Recent works show that recurrent somatic mutations target both N-terminal tails and globular histone fold domain in diverse tumor types. Oncohistones are often dominant-negative and occur at higher frequencies in tumors affecting children and adolescents. Notably, in many cases the mutations target selectively only some of the genes coding the same histone protein and are frequently associated with specific tumor types or, as documented for histone variant H3.3 in pediatric glioma, with peculiar tumors arising from specific anatomic locations. Conclusion The overview of the most recent advances suggests that the oncogenic potential of histone mutations can be exerted, together with the alteration of histone PTMs, through the destabilization of nucleosome and DNA–nucleosome interactions, as well as through the disruption of higher-order chromatin structure. However, further studies are necessary to fully elucidate the mechanism of action of oncohistones, as well as to evaluate their possible application to cancer classification, prognosis and to the identification of new therapies.
Collapse
Affiliation(s)
- Stefano Amatori
- Department of Biomolecular Sciences, Molecular Pathology Laboratory "PaoLa", University of Urbino Carlo Bo, Via Arco d'Augusto 2, 61032, Fano, PU, Italy.
| | - Simona Tavolaro
- Fredis Associazione, Via Edoardo Jenner 30, 00151, Rome, Italy
| | - Stefano Gambardella
- Department of Biomolecular Sciences, Molecular Pathology Laboratory "PaoLa", University of Urbino Carlo Bo, Via Arco d'Augusto 2, 61032, Fano, PU, Italy.,IRCCS Neuromed, Via Atinense 18, 86077, Pozzilli, IS, Italy
| | - Mirco Fanelli
- Department of Biomolecular Sciences, Molecular Pathology Laboratory "PaoLa", University of Urbino Carlo Bo, Via Arco d'Augusto 2, 61032, Fano, PU, Italy.
| |
Collapse
|
38
|
Fishbein L, Del Rivero J, Else T, Howe JR, Asa SL, Cohen DL, Dahia PLM, Fraker DL, Goodman KA, Hope TA, Kunz PL, Perez K, Perrier ND, Pryma DA, Ryder M, Sasson AR, Soulen MC, Jimenez C. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Management of Metastatic and/or Unresectable Pheochromocytoma and Paraganglioma. Pancreas 2021; 50:469-493. [PMID: 33939658 DOI: 10.1097/mpa.0000000000001792] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT This manuscript is the result of the North American Neuroendocrine Tumor Society consensus conference on the medical management and surveillance of metastatic and unresectable pheochromocytoma and paraganglioma held on October 2 and 3, 2019. The panelists consisted of endocrinologists, medical oncologists, surgeons, radiologists/nuclear medicine physicians, nephrologists, pathologists, and radiation oncologists. The panelists performed a literature review on a series of questions regarding the medical management of metastatic and unresectable pheochromocytoma and paraganglioma as well as questions regarding surveillance after resection. The panelists voted on controversial topics, and final recommendations were sent to all panel members for final approval.
Collapse
Affiliation(s)
- Lauren Fishbein
- From the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tobias Else
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - James R Howe
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Sylvia L Asa
- Department of Pathology, University Hospitals Cleveland Medical Center and University Health Network, Toronto, Case Western Reserve University, Cleveland, OH
| | - Debbie L Cohen
- Renal Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Patricia L M Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX
| | - Douglas L Fraker
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania and Abramson Cancer Center, Philadelphia, PA
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA
| | - Pamela L Kunz
- Division of Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nancy D Perrier
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Daniel A Pryma
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mabel Ryder
- Endocrine Oncology Tumor Group, Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | - Aaron R Sasson
- Division of Surgical Oncology, Department of Surgery, Stony Brook University Medical Center, Stony Brook, NY
| | - Michael C Soulen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
39
|
Prognostic value of aberrantly expressed methylation genes in human hepatocellular carcinoma. Biosci Rep 2021; 40:226463. [PMID: 32955083 PMCID: PMC7536330 DOI: 10.1042/bsr20192593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/19/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022] Open
Abstract
Objectives: To identify the prognostic value of aberrantly methylated differentially expressed genes (DEGs) in hepatocellular carcinoma (HCC) and to explore the underlying mechanisms of tumorigenesis. Methods: Gene expression profiles (GSE65372 and GSE37988) were analyzed using GEO2R to obtain aberrantly methylated DEGs. Functional enrichment analysis of screened genes was performed by the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Cytoscape software was used to analyze the PPI network and to select hub genes. Transcriptional and proteinic expression data of hub genes were obtained through UALCAN and the Human Protein Reference Database. Finally, we analyzed the prognostic value of hub genes with the Kaplan–Meier Plotter and MethSurv database. Results: In total, 24 up-hypomethylated oncogenes and 37 down-hypermethylated tumor suppressor genes (TSGs) were identified, and 8 hub genes, including 4 up-hypomethylated oncogenes (CDC5L, MERTK, RHOA and YBX1) and 4 down-hypermethylated TSGs (BCR, DFFA, SCUBE2 and TP63), were selected by PPI. Higher expression of methylated CDC5L-cg05671347, MERTK-cg08279316, RHOA-cg05657651 and YBX1-cg16306148, and lower expression of methylated BCR-cg25410636, DFFA-cg20696875, SCUBE2-cg19000089 and TP63-cg06520450, were associated with better overall survival (OS) in HCC patients. Multivariate analysis also showed they were independent prognostic factors for OS of HCC patients. Conclusions: In summary, different expression of methylated genes above mentioned were associated with better prognosis in HCC patients. Altering the methylation status of these genes may be a therapeutic target for HCC, but it should be further evaluated in clinical studies.
Collapse
|
40
|
Papathomas TG, Suurd DPD, Pacak K, Tischler AS, Vriens MR, Lam AK, de Krijger RR. What Have We Learned from Molecular Biology of Paragangliomas and Pheochromocytomas? Endocr Pathol 2021; 32:134-153. [PMID: 33433885 DOI: 10.1007/s12022-020-09658-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Recent advances in molecular genetics and genomics have led to increased understanding of the aetiopathogenesis of pheochromocytomas and paragangliomas (PPGLs). Thus, pan-genomic studies now provide a comprehensive integrated genomic analysis of PPGLs into distinct molecularly defined subtypes concordant with tumour genotypes. In addition, new embryological discoveries have refined the concept of how normal paraganglia develop, potentially establishing a developmental basis for genotype-phenotype correlations for PPGLs. The challenge for modern pathology is to translate these scientific discoveries into routine practice, which will be based largely on histopathology for the foreseeable future. Here, we review recent progress concerning the cell of origin and molecular pathogenesis of PPGLs, including pathogenetic mechanisms, genetic susceptibility and molecular classification. The current roles and tools of pathologists are considered from a histopathological perspective, including differential diagnoses, genotype-phenotype correlations and the use of immunohistochemistry in identifying hereditary predisposition and validating genetic variants of unknown significance. Current and potential molecular prognosticators are also presented with the hope that predictive molecular biomarkers will be integrated into risk stratification scoring systems to assess the metastatic potential of these intriguing neoplasms and identify potential drug targets.
Collapse
Affiliation(s)
- Thomas G Papathomas
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, UK
| | - Diederik P D Suurd
- Department of Surgical Oncology and Endocrine Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Arthur S Tischler
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston Massachusetts, USA
| | - Menno R Vriens
- Department of Surgical Oncology and Endocrine Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alfred K Lam
- School of Medicine, Griffith University, Gold Coast, QLD, Australia.
- Pathology Queensland, Gold Coast University Hospital, Gold Coast, QLD, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| | - Ronald R de Krijger
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
41
|
Histone H3G34 Mutation in Brain and Bone Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33155138 DOI: 10.1007/978-981-15-8104-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
H3G34 mutations occur in both pediatric non-brainstem high-grade gliomas (G34R/V) and giant cell tumors of bone (G34W/L). Glioblastoma patients with G34R/V mutation have a generally adverse prognosis, whereas giant cell tumors of bone are rarely metastatic benign tumors. G34 mutations possibly disrupt the epigenome by altering H3K36 modifications, which may involve attenuating the function of SETD2 at methyltransferase. H3K36 methylation change may further lead to genomic instability, dysregulated gene expression pattern, and more mutations. In this chapter, we summarize the pathological features of each mutation type in its respective cancer, as well as the potential mechanism of their disruption on the epigenome and genomic instability. Understanding each mutation type would provide a thorough background for a thorough understanding of the cancers and would bring new insights for future investigations and the development of new precise therapies.
Collapse
|
42
|
Gao Y, Ling C, Ma X, Wang H, Cui Y, Nie M, Tong A. Recurrent Germline Mutations of CHEK2 as a New Susceptibility Gene in Patients with Pheochromocytomas and Paragangliomas. Int J Endocrinol 2021; 2021:1392386. [PMID: 34630562 PMCID: PMC8497153 DOI: 10.1155/2021/1392386] [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: 07/26/2021] [Accepted: 09/22/2021] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Recently, pheochromocytomas and paragangliomas (PPGLs) have been strongly suspected as hereditary tumors, as approximately 40% of patients carry germline mutations. In the cancers where defects occur to corrupt DNA repair and facilitate tumorigenesis, a CHEK2 strong association has been observed. Therefore, the purpose of this study was to investigate the effect of CHEK2 mutations for its possible pathogenicity in PPGLs. METHODS Four patients with CHEK2 mutations were recruited, as previously detected by the whole exome sequencing. Sanger sequencing was used to verify the germline mutations as well as the loss of heterozygosities (LOHs) in their somatic DNAs. Immunohistochemistry was used to analyze the expression of CHEK2 and its downstream target p53 Ser20 (phosphorylated p53). RESULTS The average age of studied patients was 44.25 ± 11.18 years, at the time diagnosis. One patient had multiple tumors which recurred quickly, while two patients had distant metastasis. None of the patient had any relevant family history. Four germline CHEK2 mutations were identified (c.246_260del; c.715G > A; c.1008+3A > T; and c.1111C > T). All the patients were predicted to have either pathogenic or suspected pathogenic mutations. There was no LOH of CHEK2 gene in somatic DNAs found. Additionally, neither CHEK2 proteins nor its downstream target p53 Ser20 were expressed in the tumor tissues. The inactivation of CHEK2 leads to the decrease in the p53 phosphorylation, which might promote tumorigenesis. CONCLUSIONS For the first time, CHEK2 was identified as a susceptibility gene for PPGLs. However, the penetrance of CHEK2 gene with genotype-phenotype correlation needs to be investigated.
Collapse
Affiliation(s)
- Yinjie Gao
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chao Ling
- Laboratory of Clinical Genetics (Peking Union Medical College Hospital), Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaosen Ma
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Huiping Wang
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yunying Cui
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Min Nie
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Anli Tong
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
43
|
Dariane C, Goncalves J, Timsit MO, Favier J. An update on adult forms of hereditary pheochromocytomas and paragangliomas. Curr Opin Oncol 2021; 33:23-32. [PMID: 33186184 DOI: 10.1097/cco.0000000000000694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Pheochromocytomas and paragangliomas (PPGL) display a strong genetic determinism with 40% of inherited forms. The purpose of this review is to provide an update on current knowledge on adult forms of hereditary PPGL and their management. RECENT FINDINGS PPGL are genetically-driven in 70% of cases, with germline and/or somatic mutations identified in more than 20 genes. Although eight new susceptibility genes have recently emerged, mutations on SDHx genes remain the most frequent. In addition to SDHB, mutations in SLC25A11, FH and MDH2 may predispose to a metastatic disease and somatic alterations including TERT and ATRX mutations, and the differential expression on noncoding RNAs are also associated with the occurrence of metastases.The biochemical diagnosis remains the mainstay of functional PPGL and does not differ between hereditary PPGL while the choice of the best nuclear imaging approach is dictated by the tumor type and can be influenced by the presence of a germline mutation (18F-DOPA PET/CT for cluster 2 mutation and Ga-DOTATATE PET/CT for cluster 1 mutation). SUMMARY A systematic genetic testing and counselling is recommended for all PPGL patients and should lead to conservative surgery and an adapted follow up, in case of hereditary form.
Collapse
Affiliation(s)
- Charles Dariane
- Service d'urologie, Hôpital européen Georges-Pompidou, Université de Paris
| | - Judith Goncalves
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
| | - Marc-Olivier Timsit
- Service d'urologie, Hôpital européen Georges-Pompidou, Université de Paris
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
| | - Judith Favier
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, Paris, France
| |
Collapse
|
44
|
Cimmino F, Lasorsa VA, Vetrella S, Iolascon A, Capasso M. A Targeted Gene Panel for Circulating Tumor DNA Sequencing in Neuroblastoma. Front Oncol 2020; 10:596191. [PMID: 33381456 PMCID: PMC7769379 DOI: 10.3389/fonc.2020.596191] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background Liquid biopsies do not reflect the complete mutation profile of the tumor but have the potential to identify actionable mutations when tumor biopsies are not available as well as variants with low allele frequency. Most retrospective studies conducted in small cohorts of pediatric cancers have illustrated that the technology yield substantial potential in neuroblastoma. Aim The molecular landscape of neuroblastoma harbors potentially actionable genomic alterations. We aimed to study the utility of liquid biopsy to characterize the mutational landscape of primary neuroblastoma using a custom gene panel for ctDNA targeted sequencing. Methods Targeted next-generation sequencing (NGS) was performed on ctDNA of 11 patients with primary neuroblastoma stage 4. To avoid the detection of false variants, we used UMIs (unique molecular identifiers) for the library construction, increased the sequencing depth and developed ad hoc bioinformatic analyses including the hard filtering of the variant calls. Results We identified 9/11 (81.8%) patients who carry at least one pathogenic variation. The most frequently mutated genes were KMT2C (five cases), NOTCH1/2 (four cases), CREBBP (three cases), ARID1A/B (three cases), ALK (two cases), FGFR1 (two cases), FAT4 (two cases) and CARD11 (two cases). Conclusions We developed a targeted NGS approach to identify tumor-specific alterations in ctDNA of neuroblastoma patients. Our results show the reliability of our approach to generate genomic information which can be integrated with clinical and pathological data at diagnosis.
Collapse
Affiliation(s)
| | - Vito Alessandro Lasorsa
- CEINGE Biotecnologie Avanzate, Napoli, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Simona Vetrella
- Department of Pediatric Oncology, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Achille Iolascon
- CEINGE Biotecnologie Avanzate, Napoli, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Mario Capasso
- CEINGE Biotecnologie Avanzate, Napoli, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
| |
Collapse
|
45
|
Economides MP, Shah AY, Jimenez C, Habra MA, Desai M, Campbell MT. A Durable Response With the Combination of Nivolumab and Cabozantinib in a Patient With Metastatic Paraganglioma: A Case Report and Review of the Current Literature. Front Endocrinol (Lausanne) 2020; 11:594264. [PMID: 33329398 PMCID: PMC7731902 DOI: 10.3389/fendo.2020.594264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 12/04/2022] Open
Abstract
Introduction Pheochromocytomas and sympathetic paragangliomas (PPGL) are neuroendocrine catecholamine-secreting tumors that are usually localized. Metastatic disease is rare and systemic treatment consists of conventional chemotherapy and high-specific-activity iodine-131-MIBG which was approved by the FDA in 2018. Although chemotherapy combinations still have value in specific settings, the debilitating side effects of treatment with only modest benefit have limited their use. With the introduction of a new generation of targeted therapy and immunotherapy patients with metastatic PPGL may have improved therapeutic options. Areas Covered The current paper presents a case of a patient with metastatic PPGL who received multiple lines of systemic treatment. Despite progression on previous single agent cabozantinib and single agent pembrolizumab on separate clinical trials, the patient has exhibited a major response to the combination of cabozantinib and nivolumab for the past 22 months. In addition, we will review the available therapies for metastatic PPGL and discuss novel agents under clinical development. Conclusion Newer targeted therapies and immunotherapy options are under clinical development with promising results for patients with PPGL.
Collapse
Affiliation(s)
- Minas P. Economides
- Department of Internal Medicine, The University of Texas School of Health Sciences at Houston, Houston, TX, United States
| | - Amishi Y. Shah
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mouhammed A. Habra
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Monica Desai
- Department of Oncology/Hematology, Houston Methodist Cancer Center, Houston, TX, United States
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
46
|
Novel DNMT3A Germline Variant in a Patient with Multiple Paragangliomas and Papillary Thyroid Carcinoma. Cancers (Basel) 2020; 12:cancers12113304. [PMID: 33182397 PMCID: PMC7697455 DOI: 10.3390/cancers12113304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The use of next generation technologies has helped to unravel the genetics of rare inherited diseases, facilitating the discovery of new susceptibility genes. Nonetheless, the sequencing of all protein-coding genes of an individual may lead to doubtful assignments of causality for non-pathological variants, so it is mandatory to perform comprehensive studies and screening of additional patients for new mutations. Here we describe a novel DNMT3A germline variant identified by whole-exome sequencing in a patient with multiple paragangliomas and papillary thyroid carcinoma. The increased methylation of DNMT3A target genes observed in the proband’s sample points towards a gain-of-function effect of the variant, contrasting with the inactivation caused by loss-of-function alterations commonly seen in other neoplasia and in patients with Tatton-Brown-Rahman syndrome. This finding stresses the diverse molecular outcomes and suggests a heterogeneous phenotypic spectrum related to DNMT3A germline variants. Abstract Over the past few years, next generation technologies have been applied to unravel the genetics of rare inherited diseases, facilitating the discovery of new susceptibility genes. We recently found germline DNMT3A gain-of-function variants in two patients with head and neck paragangliomas causing a characteristic hypermethylated DNA profile. Here, whole-exome sequencing identifies a novel germline DNMT3A variant (p.Gly332Arg) in a patient with bilateral carotid paragangliomas, papillary thyroid carcinoma and idiopathic intellectual disability. The variant, located in the Pro-Trp-Trp-Pro (PWWP) domain of the protein involved in chromatin targeting, affects a residue mutated in papillary thyroid tumors and located between the two residues found mutated in microcephalic dwarfism patients. Structural modelling of the variant in the DNMT3A PWWP domain predicts that the interaction with H3K36me3 will be altered. An increased methylation of DNMT3A target genes, compatible with a gain-of-function effect of the alteration, was observed in saliva DNA from the proband and in one independent acute myeloid leukemia sample carrying the same p.Gly332Arg variant. Although further studies are needed to support a causal role of DNMT3A variants in paraganglioma, the description of a new DNMT3A alteration in a patient with multiple clinical features suggests a heterogeneous phenotypic spectrum related to DNMT3A germline variants.
Collapse
|
47
|
Adlard J. Multiple primary cancers (renal papillary, lymphoma and teratoma) and hepatic cysts in association with a pathogenic germline mutation in the MET gene. Fam Cancer 2020; 20:81-83. [PMID: 32686009 DOI: 10.1007/s10689-020-00196-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/15/2020] [Indexed: 11/24/2022]
Abstract
Activating germline mutations of the MET gene are associated with hereditary papillary renal cancer. This a very rare autosomal dominant condition, which is usually considered not to display a phenotype of multiple types of malignancy. However, this report describes the case of a man who has been affected with testicular teratoma, diffuse large B-cell lymphoma and multiple hepatic cysts, as well as multiple papillary renal cancers. There is good supporting evidence for roles of over-expression/activity of the HGF/MET ligand-receptor in development of these tumours, raising the possibility of other increased cancer risks associated with activating germline MET gene mutations.
Collapse
Affiliation(s)
- Julian Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, LS7 4SA, Leeds, United Kingdom.
| |
Collapse
|
48
|
Jin W. The Role of Tyrosine Kinases as a Critical Prognostic Parameter and Its Targeted Therapies in Ewing Sarcoma. Front Cell Dev Biol 2020; 8:613. [PMID: 32754598 PMCID: PMC7381324 DOI: 10.3389/fcell.2020.00613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Ewing sarcoma (ES) is a rare, highly aggressive, bone, or soft tissue-associated tumor. Although this sarcoma often responds well to initial chemotherapy, 40% of the patients develop a lethal recurrence of the disease, with death recorded in 75-80% of patients with metastatic ES within 5 years, despite receiving high-dose chemotherapy. ES is genetically well-characterized, as indicated by the EWS-FLI1 fusion protein encoded as a result of chromosomal translocation in 80-90% of patients with ES, as well as in ES-related cancer cell lines. Recently, tyrosine kinases have been identified in the pathogenesis of ES. These tyrosine kinases, acting as oncoproteins, are associated with the clinical pathogenesis, metastasis, acquisition of self-renewal traits, and chemoresistance of ES, through the activation of various intracellular signaling pathways. This review describes the recent progress related to cellular and molecular functional roles of tyrosine kinases in the progression of ES.
Collapse
Affiliation(s)
- Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon, South Korea
| |
Collapse
|
49
|
Buffet A, Burnichon N, Favier J, Gimenez-Roqueplo AP. An overview of 20 years of genetic studies in pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2020; 34:101416. [PMID: 32295730 DOI: 10.1016/j.beem.2020.101416] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Paragangliomas and pheochromocytomas (PPGL) are rare neuroendocrine tumours characterized by a strong genetic determinism. Over the past 20 years, evolution of PPGL genetics has revealed that around 40% of PPGL are genetically determined, secondary to a germline mutation in one of more than twenty susceptibility genes reported so far. More than half of the mutations occur in one of the SDHx genes (SDHA, SDHB, SDHC, SDHD, SDHAF2), which encode the different subunits and assembly protein of a mitochondrial enzyme, succinate dehydrogenase. These susceptibility genes predispose to early forms (VHL, RET, SDHD, EPAS1, DLST), syndromic (RET, VHL, EPAS1, NF1, FH), multiple (SDHD, TMEM127, MAX, DLST, MDH2, GOT2) or malignant (SDHB, FH, SLC25A11) PPGL. The discovery of a germline mutation in one of these genes changes the patient's follow-up and allows genetic screening of affected families and the presymptomatic follow-up of relatives carrying a mutation.
Collapse
Affiliation(s)
- Alexandre Buffet
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, F-75015, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, F-75015, Paris, France.
| | - Nelly Burnichon
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, F-75015, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, F-75015, Paris, France
| | - Judith Favier
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, F-75015, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, F-75015, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, F-75015, Paris, France
| |
Collapse
|
50
|
Job S, Georges A, Burnichon N, Buffet A, Amar L, Bertherat J, Bouatia-Naji N, de Reyniès A, Drui D, Lussey-Lepoutre C, Favier J, Gimenez-Roqueplo AP, Castro-Vega LJ. Transcriptome Analysis of lncRNAs in Pheochromocytomas and Paragangliomas. J Clin Endocrinol Metab 2020; 105:5611198. [PMID: 31678991 DOI: 10.1210/clinem/dgz168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
CONTEXT Pheochromocytomas and paragangliomas (PPGLs) are neuroendocrine tumors explained by germline or somatic mutations in about 70% of cases. Patients with SDHB mutations are at high risk of developing metastatic disease, yet no reliable tumor biomarkers are available to predict tumor aggressiveness. OBJECTIVE We aimed at identifying long noncoding RNAs (lncRNAs) specific for PPGL molecular groups and metastatic progression. DESIGN AND METHODS To analyze the expression of lncRNAs, we used a mining approach of transcriptome data from a well-characterized series of 187 tumor tissues. Clustering consensus analysis was performed to determine a lncRNA-based classification, and informative transcripts were validated in an independent series of 51 PPGLs. The expression of metastasis-related lncRNAs was confirmed by RT-qPCR. Receiver operating characteristic (ROC) curve analysis was used to estimate the predictive accuracy of potential markers. MAIN OUTCOME MEASURE Univariate/multivariate and metastasis-free survival (MFS) analyses were carried out for the assessment of risk factors and clinical outcomes. RESULTS Four lncRNA-based subtypes strongly correlated with mRNA expression clusters (chi-square P-values from 1.38 × 10-32 to 1.07 × 10-67). We identified one putative lncRNA (GenBank: BC063866) that accurately discriminates metastatic from benign tumors in patients with SDHx mutations (area under the curve 0.95; P = 4.59 × 10-05). Moreover, this transcript appeared as an independent risk factor associated with poor clinical outcome of SDHx carriers (log-rank test P = 2.29 × 10-05). CONCLUSION Our findings extend the spectrum of transcriptional dysregulations in PPGL to lncRNAs and provide a novel biomarker that could be useful to identify potentially metastatic tumors in patients carrying SDHx mutations.
Collapse
Affiliation(s)
- Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Adrien Georges
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Nelly Burnichon
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Genetics department, AP-HP, Hôpital européen Georges Pompidou, Paris France
| | - Alexandre Buffet
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Laurence Amar
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Hypertension unit, Paris, France
| | - Jérôme Bertherat
- INSERM, U1016, Institut Cochin, Paris, France. 10 CNRS UMR8104, Paris, France
- Rare Adrenal Cancer Network COMETE, Paris, France
| | - Nabila Bouatia-Naji
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Delphine Drui
- Service d'Endocrinologie, Diabétologie et Maladies Métaboliques, L'institut du Thorax, Centre Hospitalier Universitaire de Nantes, Hôpital Nord Laënnec, Nantes, France
| | - Charlotte Lussey-Lepoutre
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Sorbonne Université, Pitié-Salpêtrière Hospital, Department of nuclear medicine, Paris, France
| | - Judith Favier
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Genetics department, AP-HP, Hôpital européen Georges Pompidou, Paris France
- Rare Adrenal Cancer Network COMETE, Paris, France
| | - Luis Jaime Castro-Vega
- Paris University, PARCC, INSERM, Equipe labellisée par la Ligue contre le cancer, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| |
Collapse
|