1
|
de Bresser CJM, de Krijger RR. The Molecular Classification of Pheochromocytomas and Paragangliomas: Discovering the Genomic and Immune Landscape of Metastatic Disease. Endocr Pathol 2024:10.1007/s12022-024-09830-3. [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] [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
|
2
|
Freie B, Ibrahim AH, Carroll PA, Bronson RT, Augert A, MacPherson D, Eisenman RN. MAX inactivation deregulates the MYC network and induces neuroendocrine neoplasia in multiple tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614255. [PMID: 39386474 PMCID: PMC11463667 DOI: 10.1101/2024.09.21.614255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The MYC transcription factor requires MAX for DNA binding and widespread activation of gene expression in both normal and neoplastic cells. Surprisingly, inactivating mutations in MAX are associated with a subset of neuroendocrine cancers including pheochromocytoma, pituitary adenoma and small cell lung cancer. Neither the extent nor the mechanisms of MAX tumor suppression are well understood. Delet-ing Max across multiple mouse neuroendocrine tissues, we find Max inactivation alone produces pituitary adenomas while Max loss cooperates with Rb1/Trp53 loss to accelerate medullary thyroid C-cell and pituitary adenoma development. In the thyroid tumor cell lines, MAX loss triggers a striking shift in genomic occupancy by other members of the MYC network (MNT, MLX, MondoA) supporting metabolism, survival and proliferation of neoplastic neuroendocrine cells. Our work reveals MAX as a broad suppressor of neuroendocrine tumorigenesis through its ability to maintain a balance of genomic occupancies among the diverse transcription factors in the MYC network.
Collapse
Affiliation(s)
- Brian Freie
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle WA USA
| | - Ali H. Ibrahim
- Human Biology and Public Health Science Divisions, Fred Hutchinson Cancer Center, Seattle WA USA
- Present address: Department of Internal Medicine, The University of Texas Health Science Center, Houston TX USA
| | | | - Roderick T Bronson
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Arnaud Augert
- Human Biology and Public Health Science Divisions, Fred Hutchinson Cancer Center, Seattle WA USA
- Present address: Yale Cancer Center, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - David MacPherson
- Human Biology and Public Health Science Divisions, Fred Hutchinson Cancer Center, Seattle WA USA
- Department of Genome Sciences, University of Washington, Seattle WA USA
| | | |
Collapse
|
3
|
Lanzaro F, De Biasio D, Cesaro FG, Stampone E, Tartaglione I, Casale M, Bencivenga D, Marzuillo P, Roberti D. Childhood Multiple Endocrine Neoplasia (MEN) Syndromes: Genetics, Clinical Heterogeneity and Modifying Genes. J Clin Med 2024; 13:5510. [PMID: 39336996 PMCID: PMC11432259 DOI: 10.3390/jcm13185510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/15/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple endocrine neoplasia (MEN) syndromes are part of a spectrum of clinically well-defined tumor syndromes ultimately characterized by histologically similar tumors arising in patients and families with mutations in one of the following four genes: MEN1, RET, CDKN1B, and MAX. The high level of genetic and phenotypic heterogeneity has been linked to phenocopies and modifying genes, as well as unknown mechanisms that might be investigated in the future based on preclinical and translational considerations. MEN1, also known as Wermer's syndrome (OMIM *131100), is an autosomal dominant syndrome codifying for the most frequent MEN syndrome showing high penetrance due to mutations in the MEN1 gene; nevertheless, clinical manifestations vary among patients in terms of tumor localization, age of onset, and clinical aggressiveness/severity, even within the same families. This has been linked to the effect of modifying genes, as described in the review. MEN 2-2b-4 and 5 also show remarkable clinical heterogeneity. The traditional view of genetically predisposing monogenic or multifactorial disorders is no longer valid, and mandates a change in scientific focus. Phenotypes are indeed rarely consistent across genetic backgrounds and environments. In the future, understanding factors and genetic variants that control cellular functions and the expression of disease genes should provide insights into fundamental disease processes, providing implications for counseling and therapeutic and prophylactic possibilities.
Collapse
Affiliation(s)
- Francesca Lanzaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Delia De Biasio
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Francesco Giustino Cesaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Emanuela Stampone
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Immacolata Tartaglione
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Maddalena Casale
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Domenico Roberti
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| |
Collapse
|
4
|
Cetani F, Dinoi E, Pierotti L, Pardi E. Familial states of primary hyperparathyroidism: an update. J Endocrinol Invest 2024; 47:2157-2176. [PMID: 38635114 DOI: 10.1007/s40618-024-02366-7] [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: 10/25/2023] [Accepted: 03/24/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Familial primary hyperparathyroidism (PHPT) includes syndromic and non-syndromic disorders. The former are characterized by the occurrence of PHPT in association with extra-parathyroid manifestations and includes multiple endocrine neoplasia (MEN) types 1, 2, and 4 syndromes, and hyperparathyroidism-jaw tumor (HPT-JT). The latter consists of familial hypocalciuric hypercalcemia (FHH) types 1, 2 and 3, neonatal severe primary hyperparathyroidism (NSHPT), and familial isolated primary hyperparathyroidism (FIHP). The familial forms of PHPT show different levels of PHPT penetrance, developing earlier and with multiglandular involvement compared to sporadic counterpart. All these diseases exhibit Mendelian inheritance patterns, and for most of them, the genes responsible have been identified. DNA testing for predisposing mutations is helpful in index cases or in individuals with a high suspicion of the disease. Early recognition of hereditary disorders of PHPT is of great importance for the best clinical and surgical approach. Genetic testing is useful in routine clinical practice because it will also involve appropriate screening for extra-parathyroidal manifestations related to the syndrome as well as the identification of asymptomatic carriers of the mutation. PURPOSE The aim of the review is to discuss the current knowledge on the clinical and genetic profile of these disorders along with the importance of genetic testing in clinical practice.
Collapse
Affiliation(s)
- F Cetani
- Endocrine Unit 2, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - E Dinoi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L Pierotti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - E Pardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
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
|
6
|
Wachtel H, Nathanson KL. Molecular Genetics of Pheochromocytoma/Paraganglioma. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 36:100527. [PMID: 39328362 PMCID: PMC11424047 DOI: 10.1016/j.coemr.2024.100527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are neuroendocrine tumors which secrete catecholamines, causing cardiovascular compromise. While isolated tumors and locoregional disease can be treated surgically, treatment options for metastatic disease are limited, and no targeted therapies exist. Approximately 25% of PPGL are causatively associated with germline pathogenic variants, which are known risk factors for multifocal and metastatic PPGL. Knowledge of somatic driver mutations continues to evolve. Molecular classification of PPGL has identified three genomic subtypes: Cluster 1 (pseudohypoxia), Cluster 2 (kinase signaling) and Cluster 3 (Wnt-altered). This review summaries recent studies characterizing the tumor microenvironment, genomic drivers of tumorigenesis and progression, and current research on molecular targets for novel diagnostic and therapeutic strategies in PPGL.
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, PA
| | - Katherine L Nathanson
- Hospital of the University of Pennsylvania, Department of Medical Genetics, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Lian B, Lu J, Fang X, Zhang Y, Wang W, He Y, Yu H, Li F, Wang J, Chen W, Qi X. Genotype and clinical phenotype characteristics of MAX germline mutation-associated pheochromocytoma/paraganglioma syndrome. Front Endocrinol (Lausanne) 2024; 15:1442691. [PMID: 39279998 PMCID: PMC11392793 DOI: 10.3389/fendo.2024.1442691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Objective The aim of this study was to investigate the genotypic and clinical phenotypic characteristics of MAX germline mutation-associated pheochromocytoma (PCC) and paraganglioma (PGL). Methods We retrospectively analyzed the family investigation data and clinical genetic characteristics of six individuals from three independent families with PCC carrying MAX germline mutations from December 2005 to March 2024. A literature review was then conducted of the six carriers and another 103 carriers from the other 84 families with MAX germline mutations reported previously. Results There were 109 patients in 87 families with all five exons and 53 types of MAX germline mutations. p.R33* (c.97C>T; 21.1%), p.R75* (c.223C>T; 13.8%), and p.A67D (c.200C>A; 7.3%), which accounted for 42.2% of mutations detected, were the most common mutations. Moreover, 101 (92.7%) patients developed PCCs, including 59 bilateral PCCs and 42 unilateral PCCs, and 19 (18.8%) patients showed metastasis. The mean age at diagnosis was 32.8 ± 12.6 (13-80) years. The male-to-female ratio was 1.3:1. In 11 (10.9%) patients, the PCC was accompanied by chest or abdominal PGL, and one other patient had sole head and neck PGL. Nine (8.3%) patients also had functional pituitary adenomas, 11 (10.9%) developed other neuroendocrine tumors (NETs), and 7 (6.4%) presented with concomitant non-NET. Meanwhile, MAX-p.Q82Tfs*89 and p.E158A mutations are reported for the first time in this study. Conclusion MAX germline mutations may cause new types of multiple endocrine neoplasia. A comprehensive baseline assessment of neural crest cell-derived diseases is recommended for all individuals with MAX germline mutations. The risk of bilateral and metastatic PCCs should also be considered.
Collapse
Affiliation(s)
- Bijun Lian
- Laboratory Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Urology, Changhai Hospital, Navy Military Medical University, Shanghai, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Jun Lu
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Xudong Fang
- Laboratory Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yiming Zhang
- Laboratory Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Wang
- Department of Urology, Tiantai People’s Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| | - Yi He
- Department of Urology, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Hongyuan Yu
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Feiping Li
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Junwei Wang
- Department of Urology, Tiantai People’s Hospital of Zhejiang Province, Taizhou, Zhejiang, China
| | - Weiying Chen
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Xiaoping Qi
- Laboratory Department of Oncologic and Urologic Surgery, The 903rd PLA Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Enze Hospital of Hangzhou Medical College, Taizhou Enze Medical Center (Group), Taizhou, China
| |
Collapse
|
8
|
Li Q, Lan Z, Jiang Y, Wang R, Li Z, Jiang X. Validation and Evaluation of 5 Scoring Systems for Predicting Metastatic Risk in Pheochromocytoma and Paraganglioma. Am J Surg Pathol 2024; 48:855-865. [PMID: 38712603 DOI: 10.1097/pas.0000000000002238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Currently, 5 scoring systems have been proposed in the literature for predicting metastatic risk in pheochromocytoma and paraganglioma (PPGL): Pheochromocytoma of the Adrenal Gland Scaled Score (PASS), Grading System for Adrenal Pheochromocytoma and Paraganglioma (GAPP), Composite Pheochromocytoma/paraganglioma Prognostic Score (COPPS), Age, Size, Extra-adrenal location, Secretion type (ASES) score, and Size, Genetic, Age, and PASS (SGAP) model. To validate and evaluate these 5 scoring systems, we conducted a retrospective review of cases diagnosed as PPGL at the Department of Pathology, West China Hospital of Sichuan University, between January 2012 and December 2019. A total of 185 PPGL cases were included, comprising 35 cases with metastasis and 150 cases remained metastasis-free for over 8 years after surgery. The criteria of the 5 scoring systems were used for scoring and risk classification. The predictive performance of the 5 scoring systems was validated, compared, and evaluated using concordance index (C-index) and decision curve analysis (DCA). The C-indices for PASS, GAPP, and SGAP were 0.600, 0.547, and 0.547, respectively, indicating low discriminative ability. In contrast, COPPS and ASES had C-indices of 0.740 and 0.706, respectively, indicating better discriminative performance. DCA also showed that the predictive capability of COPPS was superior to that of ASES, with both outperformed PASS, while PASS had better predictive ability than GAPP and SGAP. Our analysis indicated that pathology-based scoring systems cannot accurately predict metastatic risk of PPGL. Establishing a precise prediction system requires integrating clinical, pathologic, and molecular information, using a scientific methodology for predictive factor selection and weight assessment.
Collapse
Affiliation(s)
- Qin Li
- Departments of Pathology
- Neurosurgery, West China Hospital of Sichuan University
| | - Zhigang Lan
- Department of Pathology, Chengdu Fifth People's Hospital, The Fifth People's Hospital Affiliated to Chengdu University of Traditional Chinese Medicine
| | | | | | | | - Xiaolin Jiang
- Department of Pathology, Guangyuan Central Hospital, Chengdu, Sichuan Province, China
| |
Collapse
|
9
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [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: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
10
|
Jeeyavudeen MS, Mathiyalagan N, Fernandez James C, Pappachan JM. Tumor metabolism in pheochromocytomas: clinical and therapeutic implications. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:349-373. [PMID: 38745767 PMCID: PMC11090696 DOI: 10.37349/etat.2024.00222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/27/2023] [Indexed: 05/16/2024] Open
Abstract
Pheochromocytomas and paragangliomas (PPGLs) have emerged as one of the most common endocrine tumors. It epitomizes fascinating crossroads of genetic, metabolic, and endocrine oncology, providing a canvas to explore the molecular intricacies of tumor biology. Predominantly rooted in the aberration of metabolic pathways, particularly the Krebs cycle and related enzymatic functionalities, PPGLs manifest an intriguing metabolic profile, highlighting elevated levels of oncometabolites like succinate and fumarate, and furthering cellular malignancy and genomic instability. This comprehensive review aims to delineate the multifaceted aspects of tumor metabolism in PPGLs, encapsulating genetic factors, oncometabolites, and potential therapeutic avenues, thereby providing a cohesive understanding of metabolic disturbances and their ramifications in tumorigenesis and disease progression. Initial investigations into PPGLs metabolomics unveiled a stark correlation between specific genetic mutations, notably in the succinate dehydrogenase complex (SDHx) genes, and the accumulation of oncometabolites, establishing a pivotal role in epigenetic alterations and hypoxia-inducible pathways. By scrutinizing voluminous metabolic studies and exploiting technologies, novel insights into the metabolic and genetic aspects of PPGLs are perpetually being gathered elucidating complex interactions and molecular machinations. Additionally, the exploration of therapeutic strategies targeting metabolic abnormalities has burgeoned harboring potential for innovative and efficacious treatment modalities. This review encapsulates the profound metabolic complexities of PPGLs, aiming to foster an enriched understanding and pave the way for future investigations and therapeutic innovations in managing these metabolically unique tumors.
Collapse
Affiliation(s)
| | - Navin Mathiyalagan
- Department of Medical Oncology, Nottingham University Hospitals NHS Trust, NG5 1PB Nottingham, UK
| | - Cornelius Fernandez James
- Department of Endocrinology & Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, PE21 9QS Boston, UK
| | - Joseph M. Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, PR2 9HT Preston, UK
- Faculty of Science, Manchester Metropolitan University, M15 6BH Manchester, UK
- Faculty of Biology, Medicine, and Health, The University of Manchester, M13 9PL Manchester, UK
| |
Collapse
|
11
|
Sahakian N, Castinetti F, Romanet P, Reznik Y, Brue T. Updates on the genetics of multiple endocrine neoplasia. ANNALES D'ENDOCRINOLOGIE 2024; 85:127-135. [PMID: 38325596 DOI: 10.1016/j.ando.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 02/09/2024]
Abstract
Multiple endocrine neoplasia (MEN) is a group of syndromes with a genetic predisposition to the appearance of endocrine tumors, and shows autosomal dominant transmission. The advent of molecular genetics has led to improvements in the management of MEN in terms of diagnosis, prognosis and therapy. The genetics of MEN is the subject of regular updates, which will be presented throughout this paper. MEN1, the first to be described, is associated with the MEN1 gene. MEN1 is well known in terms of the observed phenotype, with genetic analysis being conclusive in 90% of patients with a typical phenotype, but is negative in around 10% of families with MEN1. Improvement in analysis techniques and the identification of other genes responsable for phenocopies allows the resolution of some, but not all, cases, notably non-familial forms suspected to be fortuitous assocations with tumors. MEN4 is a rare phenocopy of MEN1 linked to constitutional mutations in the CDKN1B gene. Though it closely resembles the phenotype of MEN1, published data suggests the appearance of tumors is later and less frequent in MEN4. MEN2, which results from mutations in the RET oncogene, shows a strong genotype-phenotype correlation. This correlation is particularly evident in the major manifestation of MEN2, medullary thyroid carcinoma (MTC), in which disease aggressiveness is dependent on the pathogenic variant of RET. However, recent studies cast doubt on this correlation between MTC and pathogenic variant. Lastly, the recent description of families carrying a mutation in MAX, which is known to predispose to the development of pheochromocytoma and paraganglioma, and presents a phenotypic spectrum that evokes MEN, suggests the existence of another syndrome, MEN5.
Collapse
Affiliation(s)
- Nicolas Sahakian
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France.
| | - Frederic Castinetti
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France
| | - Pauline Romanet
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Laboratory of Molecular Biology, Biogenopole, Timone University Hospital, AP-HM, Marseille, France
| | - Yves Reznik
- Department of endocrinology, diabetes, metabolic disorders, University Hospital Caen, Caen, France
| | - Thierry Brue
- Aix Marseille Univ, AP-HM, Inserm, MMG, MarMaRa, Marseille, France; Department of Endocrinology, CRMR HYPO, La Conception University Hospital, AP-HM, Marseille, France
| |
Collapse
|
12
|
Giorgi RB, Aroucha PT, Favreto TM, Montero MF, Velloni JMF, Korkes I, Ferreira EN, Olivati C, Lima JV, Kater CE, Costa-Barbosa FA. Pheochromocytoma/Paraganglioma (PPGL): A Misdiagnosed Cause of Hypertension during Pregnancy. Case Rep Obstet Gynecol 2024; 2024:6655229. [PMID: 38572182 PMCID: PMC10990643 DOI: 10.1155/2024/6655229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/12/2024] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Hypertension (HT) during pregnancy is not an infrequent obstetric problem, reaching a prevalence of 5-10%. This condition is highly associated with both maternal and fetal complications if not precisely diagnosed and managed. Even though primary HT, obesity, and preeclampsia are the main causes of HT in this period, other less familiar conditions must be considered during the investigation. Pheochromocytoma and paraganglioma (PPGL) are chromaffin cell tumors that produce, store, and secrete catecholamines, leading to HT and other adrenergic manifestations. Recognition of PPGL is crucial since misdiagnosis and improper management can lead to high morbidity and mortality, particularly during pregnancy. We report on two cases of PPGL diagnosed during pregnancy with different managements. Case 1 is a 25-year-old female at 31 weeks of first pregnancy, whose severe HT and life-threatening symptoms prompted an emergency delivery without previous confirmation or medical treatment of a suspected PPGL. After confirmation, a right adrenal PPGL was surgically resected 4 months later, following 15 days of medical therapy. Case 2 is a 22-year-old female at 18 weeks of pregnancy whose symptomatic PPGL was resected in the second trimester. A next-generation sequencing panel, including 23 PPGL-related genes, found no germline pathogenic variants (GPVs) in case 1 and an exon 1-4 germinative heterozygous deletion of the MAX gene in case 2. Despite the different medical approaches, both cases had satisfactory outcomes. Although uncommon, PPGL should be considered in the differential diagnosis of HT in pregnancy since missing the diagnosis and failing to introduce appropriate and timely treatment may lead to dramatic consequences for the mother and fetus. PPGL diagnosed during reproductive age is likely to result from GPV, prompting genetic investigation and counseling.
Collapse
Affiliation(s)
- Rafael Buck Giorgi
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Division of Endocrinology, Faculty of Medical Sciences and Health, Pontific Catholic University of São Paulo, Sorocaba, Brazil
| | - Priscila Teixeira Aroucha
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Thalissa M. Favreto
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Micaela F. Montero
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Julia M. F. Velloni
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Ilana Korkes
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | | | - Caroline Olivati
- Research and Development Division, Fleury Group, São Paulo, Brazil
| | - Jose Viana Lima
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Research and Development Division, Fleury Group, São Paulo, Brazil
- Division of Endocrinology and Metabolism, Department of Medicine, Santa Casa de São Paulo, São Paulo, Brazil
| | - Claudio E. Kater
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
| | - Flavia A. Costa-Barbosa
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Federal University of Sao Paulo Medical School-EPM/Unifesp, São Paulo, Brazil
- Research and Development Division, Fleury Group, São Paulo, Brazil
| |
Collapse
|
13
|
Richter S, Bechmann N. Patient Sex and Origin Influence Distribution of Driver Genes and Clinical Presentation of Paraganglioma. J Endocr Soc 2024; 8:bvae038. [PMID: 38481600 PMCID: PMC10928507 DOI: 10.1210/jendso/bvae038] [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: 02/06/2024] [Indexed: 04/07/2024] Open
Abstract
Context Sexual and ancestral differences in driver gene prevalence have been described in many cancers but have not yet been investigated in pheochromocytoma and paraganglioma (PPGL). Objective This study aims to assess whether sex and ancestry influence prevalence of PPGL driver genes and clinical presentation. Methods We conducted a retrospective analysis of patients with PPGL considering studies from 2010 onwards that included minimal data of type of disease, sex, mutated gene, and country of origin. Additional features were recorded when available (age, tumor location, bilateral or multifocal, somatic or germline, and metastatic disease). Results We included 2162 patients: 877 in Europe and 757 in Asia. Males presented more often with germline pathogenic variants (PVs) in genes activating hypoxia pathways (P = .0006) and had more often sympathetic paragangliomas (P = .0005) and metastasis (P = .0039). On the other hand, females with PPGLs due to MAX PVs were diagnosed later than males (P = .0378) and more often developed metastasis (P = .0497). European but not Asian females presented more often with PPGLs due to PVs in genes related to kinase signaling (P = .0052), particularly RET and TMEM127. Contrary to experiences from Europe, Asian patients with PPGL due to PVs in kinase signaling genes NF1, HRAS, and FGFR1 showed a high proportion of sympathetic tumors, while European patients almost exclusively had adrenal tumors (P < .005). Conclusion Personalized management of patients with PPGL might benefit from considering sexual and ancestral differences. Further studies with better clinically aligned cohorts from various origins are required to better dissect ancestral influences on PPGL development.
Collapse
Affiliation(s)
- Susan Richter
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
14
|
English KA, Lines KE, Thakker RV. Genetics of hereditary forms of primary hyperparathyroidism. Hormones (Athens) 2024; 23:3-14. [PMID: 38038882 PMCID: PMC10847196 DOI: 10.1007/s42000-023-00508-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Primary hyperparathyroidism (PHPT), a relatively common disorder characterized by hypercalcemia with raised or inappropriately normal serum parathyroid hormone (PTH) concentrations, may occur as part of a hereditary syndromic disorder or as a non-syndromic disease. The associated syndromic disorders include multiple endocrine neoplasia types 1-5 (MEN1-5) and hyperparathyroidism with jaw tumor (HPT-JT) syndromes, and the non-syndromic forms include familial hypocalciuric hypercalcemia types 1-3 (FHH1-3), familial isolated hyperparathyroidism (FIHP), and neonatal severe hyperparathyroidism (NS-HPT). Such hereditary forms may occur in > 10% of patients with PHPT, and their recognition is important for implementation of gene-specific screening protocols and investigations for other associated tumors. Syndromic PHPT tends to be multifocal and multiglandular with most patients requiring parathyroidectomy with the aim of limiting end-organ damage associated with hypercalcemia, particularly osteoporosis, nephrolithiasis, and renal failure. Some patients with non-syndromic PHPT may have mutations of the MEN1 gene or the calcium-sensing receptor (CASR), whose loss of function mutations usually cause FHH1, a disorder associated with mild hypercalcemia and may follow a benign clinical course. Measurement of the urinary calcium-to-creatinine ratio clearance (UCCR) may help to distinguish patients with FHH from those with PHPT, as the majority of FHH patients have low urinary calcium excretion (UCCR < 0.01). Once genetic testing confirms a hereditary cause of PHPT, further genetic testing can be offered to the patients' relatives and subsequent screening can be carried out in these affected family members, which prevents inappropriate testing in normal individuals.
Collapse
Affiliation(s)
- Katherine A English
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK
| | - Kate E Lines
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| | - Rajesh V Thakker
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK.
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK.
| |
Collapse
|
15
|
Whitworth J, Armstrong R, Maher ER. Wilms tumour resulting from paternal transmission of a TRIM28 pathogenic variant-A first report. Eur J Hum Genet 2024; 32:361-364. [PMID: 38282073 PMCID: PMC10923773 DOI: 10.1038/s41431-024-01545-7] [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: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
Wilms tumour (nephroblastoma) is a renal embryonal tumour that is frequently caused by constitutional variants in a small range of cancer predisposition genes. TRIM28 has recently been identified as one such gene. Previously, observational data strongly suggested a parent of origin effect, whereby Wilms tumour only occurred following maternal inheritance of a pathogenic genetic variant. However, here we report a child with bilateral Wilms tumour who had inherited a pathogenic TRIM28 variant from their father. This finding suggests that genetic counselling for paternally inherited pathogenic variants in TRIM28 should include discussion of a potential risk of Wilms tumour.
Collapse
Affiliation(s)
- James Whitworth
- University of Cambridge Department of Medical Genetics, Box 238 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Ruth Armstrong
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Eamonn R Maher
- University of Cambridge Department of Medical Genetics, Box 238 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
- Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| |
Collapse
|
16
|
Harris EL, Roy V, Montagne M, Rose AMS, Livesey H, Reijnders MRF, Hobson E, Sansbury FH, Willemsen MH, Pfundt R, Warren D, Long V, Carr IM, Brunner HG, Sheridan EG, Firth HV, Lavigne P, Poulter JA. A recurrent de novo MAX p.Arg60Gln variant causes a syndromic overgrowth disorder through differential expression of c-Myc target genes. Am J Hum Genet 2024; 111:119-132. [PMID: 38141607 PMCID: PMC10806738 DOI: 10.1016/j.ajhg.2023.11.010] [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: 05/30/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023] Open
Abstract
Cyclin D2 (CCND2) stabilization underpins a range of macrocephaly-associated disorders through mutation of CCND2 or activating mutations in upstream genes encoding PI3K-AKT pathway components. Here, we describe three individuals with overlapping macrocephaly-associated phenotypes who carry the same recurrent de novo c.179G>A (p.Arg60Gln) variant in Myc-associated factor X (MAX). The mutation, located in the b-HLH-LZ domain, causes increased intracellular CCND2 through increased transcription but it does not cause stabilization of CCND2. We show that the purified b-HLH-LZ domain of MAXArg60Gln (Max∗Arg60Gln) binds its target E-box sequence with a lower apparent affinity. This leads to a more efficient heterodimerization with c-Myc resulting in an increase in transcriptional activity of c-Myc in individuals carrying this mutation. The recent development of Omomyc-CPP, a cell-penetrating b-HLH-LZ-domain c-Myc inhibitor, provides a possible therapeutic option for MAXArg60Gln individuals, and others carrying similar germline mutations resulting in dysregulated transcriptional c-Myc activity.
Collapse
Affiliation(s)
- Erica L Harris
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Vincent Roy
- Département de Biochimie et Génomique Fonctionnelle, PROTÉO et Institut de Pharmacologie de Sherbrooke. University of Sherbrooke, Sherbrooke, QC, Canada
| | - Martin Montagne
- Département de Biochimie et Génomique Fonctionnelle, PROTÉO et Institut de Pharmacologie de Sherbrooke. University of Sherbrooke, Sherbrooke, QC, Canada
| | - Ailsa M S Rose
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Helen Livesey
- Leeds Teaching Hospitals NHS Trust, Leeds, UK; All Wales Medical Genomics Service, NHS Wales Cardiff and Vale University Health Board, Cardiff, UK
| | - Margot R F Reijnders
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Emma Hobson
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Francis H Sansbury
- All Wales Medical Genomics Service, NHS Wales Cardiff and Vale University Health Board, Cardiff, UK
| | - Marjolein H Willemsen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Vernon Long
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ian M Carr
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Han G Brunner
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eamonn G Sheridan
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK; Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Helen V Firth
- Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK; Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Pierre Lavigne
- Département de Biochimie et Génomique Fonctionnelle, PROTÉO et Institut de Pharmacologie de Sherbrooke. University of Sherbrooke, Sherbrooke, QC, Canada.
| | - James A Poulter
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
| |
Collapse
|
17
|
Yang B, Zhang J, Wang J, Fan W, Barbier-Torres L, Yang X, Justo MAR, Liu T, Chen Y, Steggerda J, Ramani K, Lu SC, Yang H. CSNK2A1-mediated MAX phosphorylation upregulates HMGB1 and IL-6 expression in cholangiocarcinoma progression. Hepatol Commun 2023; 7:e00144. [PMID: 37347224 PMCID: PMC10289747 DOI: 10.1097/hc9.0000000000000144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND We established a novel diethylnitrosamine (DEN) -induced mouse model that reflected the progression of cholangiocarcinoma (CCA) from atypical cystic hyperplasia. METHODS BALB/c mice were administered DEN by oral gavage. Cells isolated from livers were analyzed for expression of CSNK2A1, MAX and MAX-interacting proteins. Human CCA cell lines (MzChA-1, HuCCT1), normal human cholangiocyte (H69), human hepatic stellate cells (LX-2), macrophages (RAW 264.7), and primary hepatic cells were used for cellular and molecular biology assays. RESULTS Expression of MAX, CSNK2A1, C-MYC, β-catenin, HMGB1, and IL-6 was upregulated in hepatic cells from CCA liver tissue. The half-life of MAX is higher in CCA cells, and this favors their proliferation. Overexpression of MAX increased growth, migration, and invasion of MzChA-1, whereas silencing of MAX had the opposite effect. MAX positively regulated IL-6 and HMGB1 through paracrine signaling in HepG2, LX2, and RAW cells and autocrine signaling in MzChA-1 cells. CSNK2A1-mediated MAX phosphorylation shifts MAX-MAX homodimer to C-MYC-MAX and β-catenin-MAX heterodimers and increases the HMGB1 and IL-6 promoter activities. Increase of MAX phosphorylation promotes cell proliferation, migration, invasion, and cholangiocarcinogenesis. The casein kinase 2 inhibitor CX-4945 induces cell cycle arrest and inhibits cell proliferation, migration, invasion, and carcinogenesis in MzChA-1 cells through the downregulation of CSNK2A1, MAX, and MAX-interaction proteins. CONCLUSION C-MYC-MAX and β-catenin-MAX binding to E-box site or β-catenin-MAX bound to TCFs/LEF1 enhanced HMGB1 or IL-6 promoter activities, respectively. IL-6 and HMGB1 secreted by hepatocytes, HSCs, and KCs exert paracrine effects on cholangiocytes to promote cell growth, migration, and invasion and lead to the progression of cholangiocarcinogenesis. CX-4945 provides perspectives on therapeutic strategies to attenuate progression from atypical cystic hyperplasia to cholangiocarcinogenesis.
Collapse
Affiliation(s)
- Bing Yang
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Geriatric Endocrinology and Metabolism, Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention and Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Zhang
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaohong Wang
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Wei Fan
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lucía Barbier-Torres
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Xi Yang
- Department of Geriatric Endocrinology and Metabolism, Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention and Clinical Research Center for Cardio-Cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Monica Anne R. Justo
- Department of General Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ting Liu
- Department of Gastroenterology, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Justin Steggerda
- Department of General Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Komal Ramani
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shelly C. Lu
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Heping Yang
- Department of Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
18
|
Mamedova EO, Lisina DV, Belaya ZE. [Rare forms of hereditary endocrine neoplasia: co-existence of pituitary adenoma and pheochromocytoma/paraganglioma]. PROBLEMY ENDOKRINOLOGII 2023; 69:24-30. [PMID: 37448268 DOI: 10.14341/probl13196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 07/15/2023]
Abstract
Functioning pituitary adenomas and pheochromocytomas/paragangliomas are rare in the general population. Pituitary adenomas occur in the familial setting in approximately 5% of cases, whereas pheochromocytomas/paragangliomas can be hereditary in 30-40% of cases. Hereditary syndromes associated with pituitary adenomas include multiple endocrine neoplasia types 1 and 4, familial isolated pituitary adenomas, and Carney complex. Hereditary syndromes associated with pheochromocytomas/paragangliomas and genes, mutations in which predispose to their development, are more numerous. The first clinical descriptions of the co-occurrence of pituitary adenoma and pheochromocytoma/paraganglioma in one patient date back to the mid 20th century, however delineating such a co-occurrence into a particular syndrome («3PAs» (pituitary adenoma, pheochromocytoma, paraganglioma)) was suggested only in 2015. To date, approximately 100 cases of such a co-occurrence have been described in the literature. Mutations in genes encoding subunits of succinate dehydrogenase complex II (SDHx) are revealed in the majority of cases, much less common are mutations in MAX, MEN1 and some other genes. This review summarizes the current information on the «3PAs» syndrome.
Collapse
|
19
|
Vamvoukaki R, Chrysoulaki M, Betsi G, Xekouki P. Pituitary Tumorigenesis-Implications for Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040812. [PMID: 37109772 PMCID: PMC10145673 DOI: 10.3390/medicina59040812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs), the third most common intracranial tumor, are mostly benign. However, some of them may display a more aggressive behavior, invading into the surrounding structures. While they may rarely metastasize, they may resist different treatment modalities. Several major advances in molecular biology in the past few years led to the discovery of the possible mechanisms involved in pituitary tumorigenesis with a possible therapeutic implication. The mutations in the different proteins involved in the Gsa/protein kinase A/c AMP signaling pathway are well-known and are responsible for many PitNETS, such as somatotropinomas and, in the context of syndromes, as the McCune-Albright syndrome, Carney complex, familiar isolated pituitary adenoma (FIPA), and X-linked acrogigantism (XLAG). The other pathways involved are the MAPK/ERK, PI3K/Akt, Wnt, and the most recently studied HIPPO pathways. Moreover, the mutations in several other tumor suppressor genes, such as menin and CDKN1B, are responsible for the MEN1 and MEN4 syndromes and succinate dehydrogenase (SDHx) in the context of the 3PAs syndrome. Furthermore, the pituitary stem cells and miRNAs hold an essential role in pituitary tumorigenesis and may represent new molecular targets for their diagnosis and treatment. This review aims to summarize the different cell signaling pathways and genes involved in pituitary tumorigenesis in an attempt to clarify their implications for diagnosis and management.
Collapse
Affiliation(s)
- Rodanthi Vamvoukaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Maria Chrysoulaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Grigoria Betsi
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| |
Collapse
|
20
|
Lui MS, Clemente-Gutierrez U, Skefos CM, Perrier ND. Succinate Dehydrogenase Mutations as Familial Pheochromocytoma Syndromes. Surg Oncol Clin N Am 2023; 32:289-301. [PMID: 36925186 DOI: 10.1016/j.soc.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
It is recognized that a large portion of pheochromocytoma and paraganglioma cases will have an underlying germline mutation, supporting the recommendation for universal genetic testing in all patients with PPGLs. A mutation in succinate dehydrogenase subunit B is associated with increased rates of developing synchronous and/or metachronous metastatic disease. Patients identified with this mutation require meticulous preoperative evaluation, a personalized surgical plan to minimize the risk of recurrence and tumor spread, and lifelong surveillance.
Collapse
Affiliation(s)
- Michael S Lui
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| | - Uriel Clemente-Gutierrez
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| | - Catherine M Skefos
- Clinical Cancer Genetics Program, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Nancy D Perrier
- Department of Surgical Oncology, Division of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Chevarin M, Alcantara D, Albuisson J, Collonge-Rame MA, Populaire C, Selmani Z, Baurand A, Sawka C, Bertolone G, Callier P, Duffourd Y, Jonveaux P, Bignon YJ, Coupier I, Cornelis F, Cordier C, Mozelle-Nivoix M, Rivière JB, Kuentz P, Thauvin C, Boidot R, Ghiringhelli F, O'Driscoll M, Faivre L, Nambot S. The "extreme phenotype approach" applied to male breast cancer allows the identification of rare variants of ATR as potential breast cancer susceptibility alleles. Oncotarget 2023; 14:111-125. [PMID: 36749285 PMCID: PMC9904323 DOI: 10.18632/oncotarget.28358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
In oncogenetics, some patients could be considered as "extreme phenotypes", such as those with very early onset presentation or multiple primary malignancies, unusually high numbers of cancers of the same spectrum or rare cancer types in the same parental branch. For these cases, a genetic predisposition is very likely, but classical candidate gene panel analyses often and frustratingly remains negative. In the framework of the EX2TRICAN project, exploring unresolved extreme cancer phenotypes, we applied exome sequencing on rare familial cases with male breast cancer, identifying a novel pathogenic variant of ATR (p.Leu1808*). ATR has already been suspected as being a predisposing gene to breast cancer in women. We next identified 3 additional ATR variants in a cohort of both male and female with early onset and familial breast cancers (c.7762-2A>C; c.2078+1G>A; c.1A>G). Further molecular and cellular investigations showed impacts on transcripts for variants affecting splicing sites and reduction of ATR expression and phosphorylation of the ATR substrate CHEK1. This work further demonstrates the interest of an extended genetic analysis such as exome sequencing to identify very rare variants that can play a role in cancer predisposition in extreme phenotype cancer cases unexplained by classical cancer gene panels testing.
Collapse
Affiliation(s)
- Martin Chevarin
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Unité Fonctionnelle Innovation diagnostique dans les maladies rares, laboratoire de génétique chromosomique et moléculaire, Plateau Technique de Biologie, CHU Dijon Bourgogne, Dijon, France
| | - Diana Alcantara
- Human DNA Damage Response Disorders Group, University of Sussex, Genome Damage and Stability Centre, Brighton, United Kingdom
| | - Juliette Albuisson
- Service d’Oncogénétique, Centre Georges François Leclerc, Dijon, France
- Département de biologie et pathologie des tumeurs, Centre Georges François Leclerc, Dijon, France
| | | | - Céline Populaire
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, Besançon, France
| | - Zohair Selmani
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, Besançon, France
| | - Amandine Baurand
- Service d’Oncogénétique, Centre Georges François Leclerc, Dijon, France
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
| | - Caroline Sawka
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
| | - Geoffrey Bertolone
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
| | - Patrick Callier
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Unité Fonctionnelle Innovation diagnostique dans les maladies rares, laboratoire de génétique chromosomique et moléculaire, Plateau Technique de Biologie, CHU Dijon Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Yannis Duffourd
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Philippe Jonveaux
- Laboratoire de Génétique Médicale, INSERM U954, Hôpitaux de Brabois, Vandoeuvre les Nancy, France
| | - Yves-Jean Bignon
- Laboratoire d’Oncologie Moléculaire, Centre Jean Perrin, Clermont-Ferrand, France
| | | | - François Cornelis
- Université Bordeaux, IMB, UMR 5251, Talence, France
- Service d’imagerie diagnostique et interventionnelle de l’adulte, Hôpital Pellegrin, CHU de Bordeaux, France
| | | | | | - Jean-Baptiste Rivière
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Paul Kuentz
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, Besançon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Christel Thauvin
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
| | - Romain Boidot
- Département de biologie et pathologie des tumeurs, Centre Georges François Leclerc, Dijon, France
| | - François Ghiringhelli
- Département d’oncologie médicale, INSERM LNC U1231, Centre Georges François Leclerc, Dijon, France
| | - Marc O'Driscoll
- Human DNA Damage Response Disorders Group, University of Sussex, Genome Damage and Stability Centre, Brighton, United Kingdom
| | - Laurence Faivre
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Service d’Oncogénétique, Centre Georges François Leclerc, Dijon, France
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Sophie Nambot
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Service d’Oncogénétique, Centre Georges François Leclerc, Dijon, France
- Centre de Génétique et Centre de Référence Maladies Rares Anomalies du Développement de l’Interrégion Est, Hôpital d’Enfants, CHU Dijon Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| |
Collapse
|
22
|
Charoenngam N, Mannstadt M. Primary Hyperparathyroidism in a Patient With Bilateral Pheochromocytoma and a Mutation in the Tumor Suppressor MAX. JCEM CASE REPORTS 2023; 1:luad006. [PMID: 37908276 PMCID: PMC10578369 DOI: 10.1210/jcemcr/luad006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Indexed: 11/02/2023]
Abstract
Rare heterozygous loss-of-function mutations of the MAX gene are associated with autosomal dominant hereditary pheochromocytoma-paraganglioma syndrome. In addition, evidence suggests that pathogenic MAX mutation may predispose to the development of other tumors, including endocrine and nonendocrine tumors, although the number of reported cases is small. We report a 67-year-old man with bilateral pheochromocytoma, primary hyperparathyroidism, prostate cancer, neurofibroma, and abdominal wall lipoma who tested positive for a heterozygous pathogenic germline MAX mutation. The patient has a history of bilateral norepinephrine-producing pheochromocytomas, for which he underwent left and right adrenalectomy in his 20s and 30s, respectively. His long-standing primary hyperparathyroidism was first documented when he was 40 years old and complicated by recurrent bilateral calcium oxalate nephrolithiasis and early-onset osteoporosis. Genetic testing revealed a heterozygous pathogenic deletional frameshift mutation of the exon 4 of the MAX gene (c.183_195del; p.Gln62Lysfs*104). This report, together with 3 previously reported cases, suggests that germline MAX mutation may contribute to the development of primary hyperparathyroidism and may be considered in suspected genetic forms of this disease.
Collapse
Affiliation(s)
- Nipith Charoenngam
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Medicine, Mount Auburn Hospital/Beth Israel Lahey Health, Cambridge, MA, USA
| | - Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
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
|
24
|
The Classic, the Trendy, and the Refashioned: A Primer for Pathologists on What Is New in Familial Endocrine Tumor Syndromes. Adv Anat Pathol 2023; 30:69-78. [PMID: 36136401 DOI: 10.1097/pap.0000000000000370] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Familial endocrine tumor syndromes are continuously expanding owing to the growing role of genetic testing in routine clinical practice. Pathologists are usually the first on the clinical team to encounter these syndromes at their initial presentation; thus, recognizing them is becoming more pivotal in routine pathology practice to help in properly planning management and further family testing. Our increasing knowledge about them is reflected in the newer syndromes included in the new World Health Organization classification and in the evolving discovery of new endocrine tumors and new familial associations. In many of these syndromes, the clinical features and co-occurrence of multiple neoplasia are the only clues (multiple endocrine neoplasia syndromes). In other syndromes, specific morphologic findings (pituitary blastoma and DICER1 syndrome, cribriform morular thyroid carcinoma, and AFP syndrome) and available ancillary studies (SDHB in SDH-deficient tumor syndromes) can aid pathologists. The aim of this review is to provide a primer on recent updates on familial endocrine tumor syndromes and related tumors, focusing on recent classification changes or tumor syndromes where a clearer role for pathologists is at play.
Collapse
|
25
|
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
|
26
|
Moore EC, Ioannou L, Ruseckaite R, Serpell J, Ahern S. Hereditary Endocrine Tumor Registries. J Endocr Soc 2022; 7:bvac194. [PMID: 36632485 PMCID: PMC9825730 DOI: 10.1210/jendso/bvac194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 12/24/2022] Open
Abstract
Context Endocrine neoplasia syndromes are phenotypically complex, and there is a misconception that they are universally rare. Genetic alterations are increasingly recognized; however, true prevalence is unknown. The purpose of a clinical registry is to monitor the quality of health care delivered to a specified group of patients through the collection, analysis, and reporting of relevant health-related information. This leads to improved clinical practice, decision-making, patient satisfaction, and outcome. Objective This review aims to identify, compare, and contrast active registries worldwide that capture data relevant to hereditary endocrine tumors (HETs). Methods Clinical registries were identified using a systematic approach from publications (Ovid MEDLINE, EMBASE) peer consultation, clinical trials, and web searches. Inclusion criteria were hereditary endocrine tumors, clinical registries, and English language. Exclusion criteria were institutional audits, absence of clinical data, or inactivity. Details surrounding general characteristics, funding, data fields, collection periods, and entry methods were collated. Results Fifteen registries specific for HET were shortlisted with 136 affiliated peer-reviewed manuscripts. Conclusion There are few clinical registries specific to HET. Most of these are European, and the data collected are highly variable. Further research into their effectiveness is warranted. We note the absence of an Australian registry for all HET, which would provide potential health and economic gains. This review presents a unique opportunity to harmonize registry data for HET locally and further afield.
Collapse
Affiliation(s)
- Edwina C Moore
- Correspondence: Edwina C. Moore, MBBS (HONS), BMedSci, Peninsula Private Hospital, 525 McClelland Dr, Ste 16, Langwarrin, VIC, 3199, Australia.
| | - Liane Ioannou
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| | - Rasa Ruseckaite
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| | - Jonathan Serpell
- Department of Breast, Endocrine and General Surgery, Alfred Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Susannah Ahern
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
27
|
Zethoven M, Martelotto L, Pattison A, Bowen B, Balachander S, Flynn A, Rossello FJ, Hogg A, Miller JA, Frysak Z, Grimmond S, Fishbein L, Tischler AS, Gill AJ, Hicks RJ, Dahia PLM, Clifton-Bligh R, Pacak K, Tothill RW. Single-nuclei and bulk-tissue gene-expression analysis of pheochromocytoma and paraganglioma links disease subtypes with tumor microenvironment. Nat Commun 2022; 13:6262. [PMID: 36271074 PMCID: PMC9587261 DOI: 10.1038/s41467-022-34011-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 10/10/2022] [Indexed: 01/13/2023] Open
Abstract
Pheochromocytomas (PC) and paragangliomas (PG) are rare neuroendocrine tumors associated with autonomic nerves. Here we use single-nuclei RNA-seq and bulk-tissue gene-expression data to characterize the cellular composition of PCPG and normal adrenal tissues, refine tumor gene-expression subtypes and make clinical and genotypic associations. We confirm seven PCPG gene-expression subtypes with significant genotype and clinical associations. Tumors with mutations in VHL, SDH-encoding genes (SDHx) or MAML3-fusions are characterized by hypoxia-inducible factor signaling and neoangiogenesis. PCPG have few infiltrating lymphocytes but abundant macrophages. While neoplastic cells transcriptionally resemble mature chromaffin cells, early chromaffin and neuroblast markers are also features of some PCPG subtypes. The gene-expression profile of metastatic SDHx-related PCPG indicates these tumors have elevated cellular proliferation and a lower number of non-neoplastic Schwann-cell-like cells, while GPR139 is a potential theranostic target. Our findings therefore clarify the diverse transcriptional programs and cellular composition of PCPG and identify biomarkers of potential clinical significance.
Collapse
Affiliation(s)
| | - Luciano Martelotto
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Pattison
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Blake Bowen
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiva Balachander
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Flynn
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Fernando J Rossello
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Annette Hogg
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Julie A Miller
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia.,Department of Surgery, Epworth Hospital, Richmond, VIC, Australia
| | - Zdenek Frysak
- 3rd Department of Internal Medicine - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sean Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Lauren Fishbein
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, University of Colorado, Aurora, CO, USA
| | | | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia.,NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rodney J Hicks
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Patricia L M Dahia
- Div. Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, USA
| | - Roderick Clifton-Bligh
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
28
|
Loughrey PB, Roncaroli F, Healy E, Weir P, Basetti M, Casey RT, Hunter SJ, Korbonits M. Succinate dehydrogenase and MYC-associated factor X mutations in pituitary neuroendocrine tumours. Endocr Relat Cancer 2022; 29:R157-R172. [PMID: 35938916 PMCID: PMC9513646 DOI: 10.1530/erc-22-0157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022]
Abstract
Pituitary neuroendocrine tumours (PitNETs) associated with paragangliomas or phaeochromocytomas are rare. SDHx variants are estimated to be associated with 0.3-1.8% of PitNETs. Only a few case reports have documented the association with MAX variants. Prolactinomas are the most common PitNETs occurring in patients with SDHx variants, followed by somatotrophinomas, clinically non-functioning tumours and corticotrophinomas. One pituitary carcinoma has been described. SDHC, SDHB and SDHA mutations are inherited in an autosomal dominant fashion and tumorigenesis seems to adhere to Knudson's two-hit hypothesis. SDHD and SDHAF2 mutations most commonly have paternal inheritance. Immunohistochemistry for SDHB or MAX and loss of heterozygosity analysis can support the assessment of pathogenicity of the variants. Metabolomics is promising in the diagnosis of SDHx-related disease. Future research should aim to further clarify the role of SDHx and MAX variants or other genes in the molecular pathogenesis of PitNETs, including pseudohypoxic and kinase signalling pathways along with elucidating epigenetic mechanisms to predict tumour behaviour.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
- Regional Centre for Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Division of Neuroscience and Experimental Psychology, School of Medicine, Manchester University, Manchester, UK
| | - Estelle Healy
- Department of Cellular Pathology, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Philip Weir
- Department of Neurosurgery, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Madhu Basetti
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Ruth T Casey
- Department of Endocrinology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Steven J Hunter
- Regional Centre for Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
29
|
Prinzi N, Corti F, Torchio M, Niger M, Antista M, Pagani F, Beninato T, Pulice I, Rossi RE, Coppa J, Cascella T, Giacomelli L, Di Bartolomeo M, Milione M, de Braud F, Pusceddu S. Metastatic pheochromocytomas and paragangliomas: where are we? TUMORI JOURNAL 2022; 108:526-540. [PMID: 35593402 DOI: 10.1177/03008916221078621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) can metastasize in approximately 15-20% of cases. This review discusses the available evidence on the biology and treatment of metastatic PPGLs. Chemotherapy is the first-line treatment option for this evolving and symptomatic disease. In patients with high MIBG uptake and positive PETGa-68, radiometabolic treatment may be considered. The efficacy of sunitinib has been shown in observational studies, and pembrolizumab has been evaluated in phase II clinical studies, while other agents investigated in this setting are anti-angiogenic drugs cabozantinib, dovitinib, axitinib and lenvatinib. As these agents' efficacy and safety data, alone or in combination, are scant and based on few treated patients, enrollment in clinical trials is mandatory. Future therapeutic options may be represented by DNA repair system inhibitors (such as olaparib), HIF2 inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Martina Torchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Teresa Beninato
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Iolanda Pulice
- Clinical Trial Center, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberta Elisa Rossi
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Pathophysiology and Organ Transplant, Università degli Studi di Milano, Milan, Italy
| | - Jorgelina Coppa
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso Cascella
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Massimo Milione
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy.,Oncology and Hemato-Oncology Department, Università degli Studi di Milano, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| |
Collapse
|
30
|
Hata S, Asano M, Tominaga H, Hamaguchi M, Hongo F, Usui T, Konishi E, Fukui M. Bilateral Pheochromocytoma with Germline MAX Variant without Family History. Clin Pract 2022; 12:299-305. [PMID: 35645312 PMCID: PMC9149808 DOI: 10.3390/clinpract12030035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/05/2023] Open
Abstract
Recently, the genetic background of pheochromocytomas/paragangliomas (PPGLs) has been rapidly revealed. These tumors have been referred to as the “ten percent tumor”; however, the frequency of genetic variants of PPGLs has turned out to be more common than expected. PPGLs are potentially hereditary tumors and appear clinically sporadic. Here, we report a case of bilateral pheochromocytoma (PCC) with a variant in the MYC-associated factor X (MAX) gene (c.295 + 1G > A). A male patient was diagnosed with adrenal pheochromocytoma (PCC) and underwent a left adrenalectomy at the age of 40. A new tumor in the right adrenal gland was detected at the age of 43. Urinary metanephrine and normetanephrine concentrations gradually increased. The size of the right adrenal PCC continued to increase one year after detection. Genetic testing of the peripheral blood revealed the presence of a pathogenic variant in MAX. The natural history of adrenal PCCs with the MAX variant has not yet been clarified, because the number of reported cases is not sufficient. Thus, clinicians should consider a MAX variant when they find bilateral or multiple PCCs.
Collapse
Affiliation(s)
- Shinnosuke Hata
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.H.); (H.T.); (M.H.); (M.F.)
| | - Mai Asano
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.H.); (H.T.); (M.H.); (M.F.)
- Department of Endocrinology and Metabolism, Kyoto First Red Cross Hospital, Kyoto 605-0981, Japan
| | - Hiroyuki Tominaga
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.H.); (H.T.); (M.H.); (M.F.)
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.H.); (H.T.); (M.H.); (M.F.)
| | - Fumiya Hongo
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Takeshi Usui
- Research Support Center, Shizuoka General Hospital, Shizuoka 420-8527, Japan;
- Department of Medical Genetics, Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Eiichi Konishi
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.H.); (H.T.); (M.H.); (M.F.)
| |
Collapse
|
31
|
Martinelli S, Riverso M, Mello T, Amore F, Parri M, Simeone I, Mannelli M, Maggi M, Rapizzi E. SDHB and SDHD silenced pheochromocytoma spheroids respond differently to tumour microenvironment and their aggressiveness is inhibited by impairing stroma metabolism. Mol Cell Endocrinol 2022; 547:111594. [PMID: 35149119 DOI: 10.1016/j.mce.2022.111594] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 01/11/2022] [Accepted: 02/04/2022] [Indexed: 12/18/2022]
Abstract
Germline mutations in more than 20 genes, including those encoding for the succinate dehydrogenase (SDH), predispose to rare tumours, such as pheochromocytoma/paraganglioma (PPGL). Despite encoding for the same enzymatic complex, SDHC and SDHD mutated PHEO/PGLs are generally benign, while up to 80% of SDHB mutated ones are malignant. In this study, we evaluated the different effects of tumour microenvironment on tumour cell migration/invasion, by co-culturing SDHB or SDHD silenced tumour spheroids with primary cancer-associated fibroblasts (CAFs). We observed that SDHD silenced spheroids had an intermediate migration pattern, compared to the highest migration capability of SDHB and the lowest one of the wild type (Wt) spheroids. Interestingly, we noticed that co-culturing Wt, SDHB and SDHD silenced spheroids with CAFs in low glucose (1 g/l) medium, caused a decreased migration of all the spheroids, but only for SDHB silenced ones this reduction was significant. Moreover, the collective migration, observed in high glucose (4.5 g/l) and characteristic of the SDHB silenced cells, was completely lost in low glucose. Importantly, migration could not be recovered even adding glucose (3.5 g/l) to low glucose conditioned medium. When we investigated cell metabolism, we found that low glucose concentration led to a reduction of oxygen consumption rate (OCR), basal and maximal oxidative metabolism, and ATP production only in CAFs, but not in tumour cells. These results suggest that CAFs metabolism impairment was responsible for the decreased invasion process of tumour cells, most likely preventing the release of the pro-migratory factors produced by CAFs. In conclusion, the interplay between CAFs and tumour cells is distinctive depending on the gene involved, and highlights the possibility to inhibit CAF-induced migration by impairing CAFs metabolism, indicating new potential therapeutic scenarios for medical therapy.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Maria Riverso
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Tommaso Mello
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Francesca Amore
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Matteo Parri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Irene Simeone
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine, University of Florence, Italy.
| |
Collapse
|
32
|
Tanaskovic N, Dalsass M, Filipuzzi M, Ceccotti G, Verrecchia A, Nicoli P, Doni M, Olivero D, Pasini D, Koseki H, Sabò A, Bisso A, Amati B. Polycomb group ring finger protein 6 suppresses Myc-induced lymphomagenesis. Life Sci Alliance 2022; 5:5/8/e202101344. [PMID: 35422437 PMCID: PMC9012912 DOI: 10.26508/lsa.202101344] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Max dimerizes with Mga to form the repressive complex PRC1.6; another PRC1.6 subunit, Pcgf6, suppresses Myc-induced lymphomagenesis but, unexpectedly, does so in a Mga- and PRC1.6-independent manner. Max is an obligate dimerization partner for the Myc transcription factors and for several repressors, such as Mnt, Mxd1-4, and Mga, collectively thought to antagonize Myc function in transcription and oncogenesis. Mga, in particular, is part of the variant Polycomb group repressive complex PRC1.6. Here, we show that ablation of the distinct PRC1.6 subunit Pcgf6–but not Mga–accelerates Myc-induced lymphomagenesis in Eµ-myc transgenic mice. Unexpectedly, however, Pcgf6 loss shows no significant impact on transcriptional profiles, in neither pre-tumoral B-cells, nor lymphomas. Altogether, these data unravel an unforeseen, Mga- and PRC1.6-independent tumor suppressor activity of Pcgf6.
Collapse
Affiliation(s)
| | - Mattia Dalsass
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| | | | | | | | - Paola Nicoli
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| | - Mirko Doni
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| | - Daniela Olivero
- Laboratorio Analisi Veterinarie BiEsseA, A Company of Scil Animal Care Company Srl, Milan, Italy
| | - Diego Pasini
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Haruhiko Koseki
- Laboratory of Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cellular and Molecular Medicine, Advanced Research Departments, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Arianna Sabò
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| | - Andrea Bisso
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| | - Bruno Amati
- European Institute of Oncology (IEO) - IRCCS, Milan, Italy
| |
Collapse
|
33
|
Nosé V, Gill A, Teijeiro JMC, Perren A, Erickson L. Overview of the 2022 WHO Classification of Familial Endocrine Tumor Syndromes. Endocr Pathol 2022; 33:197-227. [PMID: 35285003 DOI: 10.1007/s12022-022-09705-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2022] [Indexed: 12/16/2022]
Abstract
This review of the familial tumor syndromes involving the endocrine organs is focused on discussing the main updates on the upcoming fifth edition of the WHO Classification of Endocrine and Neuroendocrine Tumors. This review emphasizes updates on histopathological and molecular genetics aspects of the most important syndromes involving the endocrine organs. We describe the newly defined Familial Cancer Syndromes as MAFA-related, MEN4, and MEN5 as well as the newly reported pathological findings in DICER1 syndrome. We also describe the updates done at the new WHO on the syndromic and non-syndromic familial thyroid diseases. We emphasize the problem of diagnostic criteria, mention the new genes that are possibly involved in this group, and at the same time, touching upon the role of some immunohistochemical studies that could support the diagnosis of some of these conditions. As pathologists play an important role in identifying tumors within a familial cancer syndrome, we highlight the most important clues for raising the suspicious of a syndrome. Finally, we highlight the challenges in defining these entities as well as determining their clinical outcome in comparison with sporadic tumors. Instead of the usual subject review, we present the highlights of the updates on familial cancer syndromes by answering select questions relevant to practicing pathologists.
Collapse
Affiliation(s)
- Vania Nosé
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| | | | - José Manuel Cameselle Teijeiro
- Clinical University Hospital Santiago de Compostela and Medical Faculty, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
34
|
Prochownik EV, Wang H. Normal and Neoplastic Growth Suppression by the Extended Myc Network. Cells 2022; 11:747. [PMID: 35203395 PMCID: PMC8870482 DOI: 10.3390/cells11040747] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 12/20/2022] Open
Abstract
Among the first discovered and most prominent cellular oncogenes is MYC, which encodes a bHLH-ZIP transcription factor (Myc) that both activates and suppresses numerous genes involved in proliferation, energy production, metabolism and translation. Myc belongs to a small group of bHLH-ZIP transcriptional regulators (the Myc Network) that includes its obligate heterodimerization partner Max and six "Mxd proteins" (Mxd1-4, Mnt and Mga), each of which heterodimerizes with Max and largely opposes Myc's functions. More recently, a second group of bHLH-ZIP proteins (the Mlx Network) has emerged that bears many parallels with the Myc Network. It is comprised of the Myc-like factors ChREBP and MondoA, which, in association with the Max-like member Mlx, regulate smaller and more functionally restricted repertoires of target genes, some of which are shared with Myc. Opposing ChREBP and MondoA are heterodimers comprised of Mlx and Mxd1, Mxd4 and Mnt, which also structurally and operationally link the two Networks. We discuss here the functions of these "Extended Myc Network" members, with particular emphasis on their roles in suppressing normal and neoplastic growth. These roles are complex due to the temporal- and tissue-restricted expression of Extended Myc Network proteins in normal cells, their regulation of both common and unique target genes and, in some cases, their functional redundancy.
Collapse
Affiliation(s)
- Edward V. Prochownik
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
- The Department of Microbiology and Molecular Genetics, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
- The Hillman Cancer Center of UPMC, Pittsburgh, PA 15224, USA
- The Pittsburgh Liver Research Center, Pittsburgh, PA 15224, USA
| | - Huabo Wang
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
| |
Collapse
|
35
|
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
|
36
|
Luque J, Mendes I, Gómez B, Morte B, Heredia ML, Herreras E, Corrochano V, Bueren J, Gallano P, Artuch R, Fillat C, Pérez‐Jurado LA, Montoliu L, Carracedo Á, Millán JM, Webb SM, Palau F, Lapunzina P, Aguado C, Aguado C, Albiñana V, Alías L, Almoguera B, Alonso J, Alonso‐Ferreira V, Alvarez‐Mora MI, Alvarez‐Mora MI, Antiñolo G, Arbones ML, Arenas J, Arjona E, Armangue T, Armstrong J, Arnedo M, Artuch R, Masó AA, Avila‐Fernandez A, Ayuso C, Badell I, Badenas C, Baeza ML, Baiget M, Balcells S, Ballesta‐Martínez MJ, Barahona M, Barros F, Bartoccioni PC, Bayona‐Bafaluy MP, Sanz SB, Bernabéu C, Bernal S, Blanco‐Kelly F, Blázquez A, Bodoy S, Bogliolo M, Borralleras C, Borrego S, Botella LM, Pieri FB, Bovolenta P, Bravo‐Gil N, Brea A, Bueno‐Lozano G, Bueren J, Bustamante A, Caballero T, Camacho‐Macorra C, Cámara Y, Camats‐Tarruella N, Barrio ÁC, Campuzano V, Cantarero L, Cantó J, Caparrós‐Martín JA, Cardellach F, Carmona R, Carracedo Á, Carretero M, Casado M, Casado JA, Casasnovas C, Cascón A, Casino P, Castaño L, Castilla‐Vallmanya L, Catala A, Cayuela ML, Cediel R, Cervera J, Codina‐Solà M, Contreras J, Cormand B, Corominas R, Corral J, Corrochano V, Cortés‐Rodríguez A, Corton M, Costa‐Roger M, Cozar M, Crespo I, Crispi F, Cruz R, Cuezva JM, Cuscó I, Dalmau J, Cima S, Luna S, De Luna N, Oyarzabal Sanz A, Campo M, Castillo I, Molina LDP, Pozo ÁD, Río M, Delmiro A, Desviat LR, Dierssen M, Domínguez‐González C, Domínguez‐Ruiz M, Dopazo J, Errasti E, Escámez MJ, Estañ MC, Esteban J, Estévez R, Ezquieta B, Fernández L, Fernández A, Fernández‐Cancio M, Fernàndez‐Castillo N, Jose PF, Fillat C, Fons C, Fort J, Fourcade S, Fraga MF, Gallano P, Gallardo E, García M, García‐Arumí E, García‐Bravo M, García‐Cazorla A, García‐Consuegra I, Garcia‐Garcia FJ, García‐García G, García‐Giménez JL, Garcia‐Gimeno MA, García‐Miñaur S, García‐Redondo A, García‐Silva MT, García‐Villoria J, Santiago FG, Garrabou G, Garrido G, Garrido‐Pérez N, Gaztambide S, Gil‐Campos M, Giroud‐Gerbetant J, Glover G, Gómez B, Gómez‐Puertas P, Gonzalez‐Cabo P, Gonzalez‐Casacuberta I, Pozo MG, González‐Quereda L, González‐Quintana A, Gort L, Gougeard N, Gratacos E, Grau JM, Grinberg D, Güenechea G, Guerrero R, Guillén‐Navarro E, Guitart‐Mampel M, Gutiérrez‐Arumí A, Heath K, Heredia M, Hernández‐Chico C, Herreras E, Hoenicka J, Homs A, Jimenez‐Estrada JA, Jimenez‐Mallebrera C, Jou C, Juarez‐Flores DL, Lapunzina P, Larcher F, Lasa A, Lassaletta L, Latorre‐Pellicer A, Linares D, Llacer JL, Llames S, Lopez‐Gallardo E, López‐Laso E, López‐Lera A, Lopez‐Lopez D, López‐Sánchez M, Heredia ML, Granados EL, Lorda‐Sanchez I, Lozano ML, Luque J, Madrigal I, García CM, Mansilla E, Marco‐Marín C, Marfany G, Marina A, Martí R, Martí S, Martin Y, Martín MA, Martín‐Hernandez E, Martin‐Merida I, Martínez R, Martínez‐Azorín F, Martinez‐Delgado B, Martínez‐Gil N, Martínez‐Glez VM, Martínez‐Momblán MA, Martínez‐Romero MC, Fernández PM, Santamaría LM, Martorell L, Meade P, Meana Á, Medina MÁ, Mendes I, Méndez‐Vidal C, Millán JM, Minguez P, Minguillón J, Mirra S, Molla B, Moltó E, Montero R, Montoliu L, Montoya J, Morán M, Moren C, Moreno M, Moreno JC, Moreno‐Galdó A, Moreno‐Pelayo MÁ, Mori MA, Morin M, Morte B, Mulero V, Muñoz‐Pujol G, Murillas R, Murillo‐Cuesta S, Nascimento A, Navarro S, Navas P, Nevado J, Nicolas A, Nieto MÁ, O’Callaghan M, Olavarrieta L, Ormazabal A, Ortiz‐Romero P, Osorio A, Páez D, Palacín M, Palacios‐Verdú MG, Palau F, Palencia‐Campos A, Pallardó FV, Palomares M, Peña‐Chilet M, Pérez B, Perez‐Florido J, Pérez‐García D, Perez‐Jimenez E, Pérez‐Jurado LA, Perkins JR, Perona R, Pie J, Pinós T, Pinto S, Potrony M, Puig S, Puig‐Butille JA, Puisac B, Pujol R, Pujol A, Quintana Ó, Rabionet R, Ramos FJ, Ranea JAG, Reina‐Castillón J, Resmini E, Ribes A, Rica I, Richard E, Riera P, Río P, Riveiro‐Alvarez R, Rivera J, Rivera‐Barahona A, Robledo M, Rodriguez‐Aguilera JC, Rosa LR, Rodríguez‐Palmero A, Rodriguez‐Pombo P, Rodriguez‐Revenga L, Rodríguez‐Santiago B, Rodríguez‐Sureda V, Alba MR, Cordoba SR, Romá‐Mateo C, Rubio V, Ruiz Á, Ruiz M, Ruiz‐Arenas C, Ruiz‐Perez VL, Ruiz‐Pesini E, Ruiz‐Ponte C, Rullo J, Sabater L, Salazar J, Salido E, Sanchez‐Jimeno C, Cuesta AMS, Soler MJS, Santacatterina F, Santamarina M, Santos A, Santos‐Ocaña C, Simarro FS, Sanz P, Sastre L, Schlüter A, Segovia JC, Segura‐Puimedon M, Seoane P, Serra‐Juhe C, Serrano M, Serratosa JM, Sevilla T, Surrallés J, Tahsin‐Swafiri S, Tell‐Martí G, Tenorio‐Castaño JA, Tizzano E, Tobias E, Tort F, Trujillano L, Trujillo‐Tiebas MJ, Ugalde C, Ugarteburu O, Urreizti R, Urrutia I, Valencia M, Vallcorba P, Vallespín E, Varela‐Nieto I, Vega A, Vélez‐Santamaria V, Vílchez JJ, Villa O, Villamar M, Webb SM, Zubeldia JM, Zurita O. CIBERER: Spanish National Network for Research on Rare Diseases: a highly productive collaborative initiative. Clin Genet 2022; 101:481-493. [PMID: 35060122 PMCID: PMC9305285 DOI: 10.1111/cge.14113] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/30/2022]
Abstract
CIBER (Center for Biomedical Network Research; Centro de Investigación Biomédica En Red) is a public national consortium created in 2006 under the umbrella of the Spanish National Institute of Health Carlos III (ISCIII). This innovative research structure comprises 11 different specific areas dedicated to the main public health priorities in the National Health System. CIBERER, the thematic area of CIBER focused on rare diseases (RDs) currently consists of 75 research groups belonging to universities, research centers, and hospitals of the entire country. CIBERER's mission is to be a center prioritizing and favoring collaboration and cooperation between biomedical and clinical research groups, with special emphasis on the aspects of genetic, molecular, biochemical, and cellular research of RDs. This research is the basis for providing new tools for the diagnosis and therapy of low‐prevalence diseases, in line with the International Rare Diseases Research Consortium (IRDiRC) objectives, thus favoring translational research between the scientific environment of the laboratory and the clinical setting of health centers. In this article, we intend to review CIBERER's 15‐year journey and summarize the main results obtained in terms of internationalization, scientific production, contributions toward the discovery of new therapies and novel genes associated to diseases, cooperation with patients' associations and many other topics related to RD research.
Collapse
Affiliation(s)
- Juan Luque
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Ingrid Mendes
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Beatriz Gómez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Beatriz Morte
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Miguel López Heredia
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Enrique Herreras
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Virginia Corrochano
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
| | - Juan Bueren
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS‐FJD), Madrid Spain
| | - Pía Gallano
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Genetics Department, Hospital de la Santa Creu i Sant Pau Barcelona Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), Barcelona Spain
- Universitat Autònoma de Barcelona Barcelona Spain
| | - Rafael Artuch
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu Barcelona Spain
| | - Cristina Fillat
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona Spain
- Universitat de Barcelona Barcelona Spain
| | - Luis A. Pérez‐Jurado
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Department of Experimental and Health Sciences Universitat Pompeu Fabra (UPF), Barcelona Spain
- Genetics Service, Hospital del Mar Barcelona Spain
- Hospital del Mar Research Institute (IMIM), Barcelona Spain
| | - Lluis Montoliu
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB‐CSIC), Madrid Spain
| | - Ángel Carracedo
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Grupo de Medicina Xenómica, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela Santiago de Compostela Spain
- Fundación Pública Galega de Medicina Xenómica (SERGAS), IDIS Santiago de Compostela Spain
| | - José M. Millán
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Unidad de Genética, Hospital Universitario y Politécnico La Fe Valencia Spain
- Biomedicina Molecular Celular y Genómica, Instituto Investigación Sanitaria La Fe Valencia Spain
| | - Susan M. Webb
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Hospital S Pau, Dept Medicine/Endocrinology, IIB‐Sant Pau, Research Center for Pituitary Diseases Barcelona Spain
- Departamento de Medicina Universitat Autònoma de Barcelona Barcelona Spain
| | - Francesc Palau
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- Department of Genetic and Molecular Medicine, Hospital Sant Joan de Deu Barcelona Spain
- Laboratory of Neurogenetics and Molecular Medicine ‐ IPER, Institut de Recerca Sant Joan de Déu Barcelona Spain
- Institute of Medicine & Dermatology, Hospital Clínic Barcelona Spain
- Division of Pediatrics University of Barcelona School of Medicine Barcelona Spain
| | - Pablo Lapunzina
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III Madrid Spain
- INGEMM‐Instituto de Genética Médica y Molecular, Hospital Universitario La Paz Madrid Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPAZ), Madrid Spain
- ERN‐ITHACA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Steinman JR, Thomas MH, McCartney CR, Padia SH. A novel pathogenic variant in MAX-Associated pheochromocytoma. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2021. [DOI: 10.1016/j.jecr.2021.100101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
38
|
Реброва ДВ, Ворохобина НВ, Имянитов ЕН, Русаков ВФ, Краснов ЛМ, Слепцов ИВ, Черников РА, Федоров ЕА, Семенов АА, Чинчук ИК, Саблин ИВ, Алексеев МА, Кулешов ОВ, Федотов ЮН. [Clinical and laboratory features of hereditary pheochromocytoma and paraganglioma]. PROBLEMY ENDOKRINOLOGII 2021; 68:8-17. [PMID: 35262293 PMCID: PMC9761867 DOI: 10.14341/probl12834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023]
Abstract
The widespread introduction of genetic testing in recent years has made it possible to determine that more than a third of cases of pheochromocytomas and paragangliomas (PPPGs) are caused by germline mutations. Despite the variety of catecholamine-producing tumors manifestations, there is a sufficient number of clinical and laboratory landmarks that suggest a hereditary genesis of the disease and even a specific syndrome. These include a family history, age of patient, presence of concomitant conditions, and symptoms of the disease. Considering that each of the mutations is associated with certain diseases that often determine tactics of treatment and examination of a patient, e.g. high risk of various malignancies. Awareness of the practitioner on the peculiarities of the course of family forms of PPPGs will allow improving the tactics of managing these patients.The article provides up-to-date information on the prevalence of hereditary PPPGs. The modern views on the pathogenesis of the disease induced by different mutations are presented. The main hereditary syndromes associated with PPPGs are described, including multiple endocrine neoplasia syndrome type 2A and 2B, type 1 neurofibromatosis, von Hippel-Lindau syndrome, hereditary paraganglioma syndrome, as well as clinical and laboratory features of the tumor in these conditions. The main positions on the necessity of genetic screening in patients with PPPGs are given.
Collapse
Affiliation(s)
- Д. В. Реброва
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - Н. В. Ворохобина
- Северо-Западный государственный медицинский университет им. И.И. Мечникова
| | - Е. Н. Имянитов
- Национальный медицинский исследовательский центр онкологии им. Н.Н. Петрова
| | - В. Ф. Русаков
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - Л. М. Краснов
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - И. В. Слепцов
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - Р. А. Черников
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - Е. А. Федоров
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - А. А. Семенов
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - И. К. Чинчук
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - И.. В. Саблин
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - М. А. Алексеев
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - О. В. Кулешов
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| | - Ю. Н. Федотов
- Санкт-Петербургский государственный университет, Клиника высоких медицинских технологий им. Н.И. Пирогова
| |
Collapse
|
39
|
Kumar S, Lila AR, Memon SS, Sarathi V, Patil VA, Menon S, Mittal N, Prakash G, Malhotra G, Shah NS, Bandgar TR. Metastatic cluster 2-related pheochromocytoma/paraganglioma: a single-center experience and systematic review. Endocr Connect 2021; 10:1463-1476. [PMID: 34662294 PMCID: PMC8630763 DOI: 10.1530/ec-21-0455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/18/2021] [Indexed: 11/24/2022]
Abstract
Risk of metastatic disease in the cluster 2-related pheochromocytoma/paraganglioma (PPGL) is low. In MEN2 patients, identification of origin of metastases from pheochromocytoma (PCC) or medullary thyroid carcinoma (MTC) is challenging as both are of neuroendocrine origin. We aim to describe our experience and perform a systematic review to assess prevalence, demographics, biochemistry, diagnostic evaluation, management, and predictors of cluster 2-related metastatic PPGL. Retrospective analysis of 3 cases from our cohort and 43 cases from world literature was done. For calculation of prevalence, all reported patients (n = 3063) of cluster 2 were included. We found that the risk of metastasis in cluster 2-related PPGL was 2.6% (2% in RET, 5% in NF1, 4.8% in TMEM127 and 16.7% in MAX variation). In metastatic PCC in MEN2, median age was 39 years, bilateral tumors were present in 71% and median tumor size was 9.7 cm (range 4-19) with 43.5% mortality. All patients had a primary tumor size ≥4 cm. Origin of primary tumor was diagnosed by histopathology of metastatic lesion in 11 (57.9%), 131I-MIBG scan in 6 (31.6%), and selective venous sampling and CT in 1 (5.3%) patient each. In subgroup of neurofibromatosis 1 (NF1), median age was 46 years (range 14-59) with median tumor size 6 cm and 57% mortality. To conclude, the risk of metastatic disease in cluster 2-related PPGL is low, being especially high in tumors with size ≥4 cm and associated with high mortality. One-third patients of NF1 with metastatic PPGL had presented in second decade of life. Long-term studies are needed to formulate management recommendations.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Anurag Ranjan Lila
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Saba Samad Memon
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, India
| | - Virendra A Patil
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Neha Mittal
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Gagan Prakash
- Department of Uro-oncology, Tata Memorial Hospital, Mumbai, India
| | - Gaurav Malhotra
- Department of Nuclear Medicine, Bhabha Atomic Research Centre, Mumbai, India
| | - Nalini S Shah
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Tushar R Bandgar
- Department of Endocrinology, Seth G S Medical College & KEM Hospital, Mumbai, India
- Correspondence should be addressed to T R Bandgar:
| |
Collapse
|
40
|
Jiménez Martín O, Schlosser A, Furtwängler R, Wegert J, Gessler M. MYCN and MAX alterations in Wilms tumor and identification of novel N-MYC interaction partners as biomarker candidates. Cancer Cell Int 2021; 21:555. [PMID: 34689785 PMCID: PMC8543820 DOI: 10.1186/s12935-021-02259-2] [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: 06/23/2021] [Accepted: 10/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Wilms tumor (WT) is the most common renal tumor in childhood. Among others, MYCN copy number gain and MYCN P44L and MAX R60Q mutations have been identified in WT. MYCN encodes a transcription factor that requires dimerization with MAX to activate transcription of numerous target genes. MYCN gain has been associated with adverse prognosis in different childhood tumors including WT. The MYCN P44L and MAX R60Q mutations, located in either the transactivating or basic helix-loop-helix domain, respectively, are predicted to be damaging by different pathogenicity prediction tools, but the functional consequences remain to be characterized. METHODS We screened a large cohort of unselected WTs for MYCN and MAX alterations. Wild-type and mutant protein function were characterized biochemically, and we analyzed the N-MYC protein interactome by mass spectrometric analysis of N-MYC containing protein complexes. RESULTS Mutation screening revealed mutation frequencies of 3% for MYCN P44L and 0.9% for MAX R60Q that are associated with a higher risk of relapse. Biochemical characterization identified a reduced transcriptional activation potential for MAX R60Q, while the MYCN P44L mutation did not change activation potential or protein stability. The protein interactome of N-MYC-P44L was likewise not altered as shown by mass spectrometric analyses of purified N-MYC complexes. Nevertheless, we could identify a number of novel N-MYC partner proteins, e.g. PEG10, YEATS2, FOXK1, CBLL1 and MCRS1, whose expression is correlated with MYCN in WT samples and several of these are known for their own oncogenic potential. CONCLUSIONS The strongly elevated risk of relapse associated with mutant MYCN and MAX or elevated MYCN expression corroborates their role in WT oncogenesis. Together with the newly identified co-expressed interactors they expand the range of potential biomarkers for WT stratification and targeting, especially for high-risk WT.
Collapse
Affiliation(s)
- Ovidio Jiménez Martín
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, 97074, Wuerzburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Wuerzburg University, 97078, Wuerzburg, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Oncology Und Hematology, Saarland University Hospital, 66421, Homburg, Saar, Germany
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, 97074, Wuerzburg, Germany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, 97074, Wuerzburg, Germany. .,Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97078, Wuerzburg, Germany.
| |
Collapse
|
41
|
Liaño-Pons J, Arsenian-Henriksson M, León J. The Multiple Faces of MNT and Its Role as a MYC Modulator. Cancers (Basel) 2021; 13:4682. [PMID: 34572909 PMCID: PMC8465425 DOI: 10.3390/cancers13184682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
MNT is a crucial modulator of MYC, controls several cellular functions, and is activated in most human cancers. It is the largest, most divergent, and most ubiquitously expressed protein of the MXD family. MNT was first described as a MYC antagonist and tumor suppressor. Indeed, 10% of human tumors present deletions of one MNT allele. However, some reports show that MNT functions in cooperation with MYC by maintaining cell proliferation, promoting tumor cell survival, and supporting MYC-driven tumorigenesis in cellular and animal models. Although MAX was originally considered MNT's obligate partner, our recent findings demonstrate that MNT also works independently. MNT forms homodimers and interacts with proteins both outside and inside of the proximal MYC network. These complexes are involved in a wide array of cellular processes, from transcriptional repression via SIN3 to the modulation of metabolism through MLX as well as immunity and apoptosis via REL. In this review, we discuss the present knowledge of MNT with a special focus on its interactome, which sheds light on the complex and essential role of MNT in cell biology.
Collapse
Affiliation(s)
- Judit Liaño-Pons
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, SE-171 65 Stockholm, Sweden;
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, SE-171 65 Stockholm, Sweden;
| | - Javier León
- Departmento de Biología Molecular and Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, 39011 Santander, Spain;
| |
Collapse
|
42
|
Llabata P, Torres-Diz M, Gomez A, Tomas-Daza L, Romero OA, Grego-Bessa J, Llinas-Arias P, Valencia A, Esteller M, Javierre BM, Zhang X, Sanchez-Cespedes M. MAX mutant small-cell lung cancers exhibit impaired activities of MGA-dependent noncanonical polycomb repressive complex. Proc Natl Acad Sci U S A 2021; 118:e2024824118. [PMID: 34493659 PMCID: PMC8449313 DOI: 10.1073/pnas.2024824118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
The MYC axis is disrupted in cancer, predominantly through activation of the MYC family oncogenes but also through inactivation of the MYC partner MAX or of the MAX partner MGA. MGA and MAX are also members of the polycomb repressive complex, ncPRC1.6. Here, we use genetically modified MAX-deficient small-cell lung cancer (SCLC) cells and carry out genome-wide and proteomics analyses to study the tumor suppressor function of MAX. We find that MAX mutant SCLCs have ASCL1 or NEUROD1 or combined ASCL1/NEUROD1 characteristics and lack MYC transcriptional activity. MAX restitution triggers prodifferentiation expression profiles that shift when MAX and oncogenic MYC are coexpressed. Although ncPRC1.6 can be formed, the lack of MAX restricts global MGA occupancy, selectively driving its recruitment toward E2F6-binding motifs. Conversely, MAX restitution enhances MGA occupancy to repress genes involved in different functions, including stem cell and DNA repair/replication. Collectively, these findings reveal that MAX mutant SCLCs have either ASCL1 or NEUROD1 or combined characteristics and are MYC independent and exhibit deficient ncPRC1.6-mediated gene repression.
Collapse
Affiliation(s)
- Paula Llabata
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Manuel Torres-Diz
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, 08908 Barcelona, Spain
| | - Antonio Gomez
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035 Barcelona, Spain
| | - Laureano Tomas-Daza
- 3D Chromatin Organization Group, Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Octavio A Romero
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Joaquim Grego-Bessa
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, 08908 Barcelona, Spain
| | - Pere Llinas-Arias
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Alfonso Valencia
- Computational Biology Life Sciences Group, Barcelona Supercomputing Centre, 08034 Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer, 28029 Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain
| | - Biola M Javierre
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035 Barcelona, Spain
| | - Xiaoyang Zhang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | | |
Collapse
|
43
|
Liu Z, Ma J, Jimenez C, Zhang M. Pheochromocytoma: A Clinicopathologic and Molecular Study of 390 Cases From a Single Center. Am J Surg Pathol 2021; 45:1155-1165. [PMID: 34280940 DOI: 10.1097/pas.0000000000001768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pheochromocytomas are rare neuroendocrine tumors arising from chromaffin cells in the adrenal medulla. They may occur sporadically or in the context of hereditary syndromes. All pheochromocytomas are considered to have malignant potential (defined as risk of metastasis, not local invasion). The use of grading systems with incorporated clinical and histopathologic parameters can help but not definitively predict the metastatic potential of pheochromocytomas. The recent discovery of susceptibility genes provided new insights into the pathogenesis and introduced additional approaches to estimate the metastatic risk of pheochromocytoma. However, the prevalence of these genetic signatures in pheochromocytomas has yet to be fully addressed. Therefore, in the present study, we retrospectively reviewed cases of pheochromocytoma from 1980 to 2018 in the archives of our institution. Three hundred ninety cases were identified, and their clinicopathologic characteristics and genetic statuses were analyzed. About 25% of the cases had metastases, which were more common in older patients (median, 49 y) than in younger ones. Univariate and multivariate analyses revealed that older age, Hispanic ethnicity, metastasis, and large primary tumor size were markedly associated with poor overall survival. In contrast, family history of pheochromocytoma, lack of symptoms, and bilateral adrenal involvement were associated with better survival. About 37% of the pheochromocytomas were associated with inherited syndromes. About 52% of tested patients had pathogenic mutations of pheochromocytoma susceptibility genes. Of these, succinate dehydrogenase B gene mutation had the strongest association with metastasis. These data support that genetic testing should be offered to all patients with pheochromocytoma.
Collapse
Affiliation(s)
| | | | - Camilo Jimenez
- Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
44
|
Casey R, Neumann HPH, Maher ER. Genetic stratification of inherited and sporadic phaeochromocytoma and paraganglioma: implications for precision medicine. Hum Mol Genet 2021; 29:R128-R137. [PMID: 33059362 DOI: 10.1093/hmg/ddaa201] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades advances in genomic technologies have transformed knowledge of the genetic basis of phaeochromocytoma and paraganglioma (PPGL). Though traditional teaching suggested that inherited cases accounted for only 10% of all phaeochromocytoma diagnosis, current estimates are at least three times this proportion. Inherited PPGL is a highly genetically heterogeneous disorder but the most frequently results from inactivating variants in genes encoding subunits of succinate dehydrogenase. Expanding knowledge of the genetics of PPGL has been translated into clinical practice by the provision of widespread testing for inherited PPGL. In this review, we explore how the molecular stratification of PPGL is being utilized to enable more personalized strategies for investigation, surveillance and management of affected individuals and their families. Translating recent genetic research advances into clinical service can not only bring benefits through more accurate diagnosis and risk prediction but also challenges when there is a suboptimal evidence base for the clinical consequences or significance of rare genotypes. In such cases, clinical, biochemical, pathological and functional imaging assessments can all contribute to more accurate interpretation and clinical management.
Collapse
Affiliation(s)
- Ruth Casey
- Department of Medical Genetics, University of Cambridge, Cambridge, CB2 0QQ, UK.,NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK.,Department of Endocrinology, Cambridge University Hospital Foundation Trust, Cambridge CB2 0QQ, UK
| | - Hartmut P H Neumann
- Section for Preventive Medicine, Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Germany
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge, CB2 0QQ, UK.,NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| |
Collapse
|
45
|
Sarkadi B, Liko I, Nyiro G, Igaz P, Butz H, Patocs A. Analytical Performance of NGS-Based Molecular Genetic Tests Used in the Diagnostic Workflow of Pheochromocytoma/Paraganglioma. Cancers (Basel) 2021; 13:4219. [PMID: 34439371 PMCID: PMC8392134 DOI: 10.3390/cancers13164219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/30/2022] Open
Abstract
Next Generation Sequencing (NGS)-based methods are high-throughput and cost-effective molecular genetic diagnostic tools. Targeted gene panel and whole exome sequencing (WES) are applied in clinical practice for assessing mutations of pheochromocytoma/paraganglioma (PPGL) associated genes, but the best strategy is debated. Germline mutations of at the least 18 PPGL genes are present in approximately 20-40% of patients, thus molecular genetic testing is recommended in all cases. We aimed to evaluate the analytical and clinical performances of NGS methods for mutation detection of PPGL-associated genes. WES (three different library preparation and bioinformatics workflows) and an in-house, hybridization based gene panel (endocrine-onco-gene-panel- ENDOGENE) was evaluated on 37 (20 WES and 17 ENDOGENE) samples with known variants. After optimization of the bioinformatic workflow, 61 additional samples were tested prospectively. All clinically relevant variants were validated with Sanger sequencing. Target capture of PPGL genes differed markedly between WES platforms and genes tested. All known variants were correctly identified by all methods, but methods of library preparations, sequencing platforms and bioinformatical settings significantly affected the diagnostic accuracy. The ENDOGENE panel identified several pathogenic mutations and unusual genotype-phenotype associations suggesting that the whole panel should be used for identification of genetic susceptibility of PPGL.
Collapse
Affiliation(s)
- Balazs Sarkadi
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
| | - Istvan Liko
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Bionics Innovation Center, H-1089 Budapest, Hungary;
| | - Gabor Nyiro
- Bionics Innovation Center, H-1089 Budapest, Hungary;
- MTA-SE Molecular Medicine Research Group, Eotvos Lorand Research Network, H-1083 Budapest, Hungary;
| | - Peter Igaz
- MTA-SE Molecular Medicine Research Group, Eotvos Lorand Research Network, H-1083 Budapest, Hungary;
- Department of Endocrinology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Henriett Butz
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Department of Laboratory Medicine, Semmelweis University, H-1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, H-1122 Budapest, Hungary
| | - Attila Patocs
- MTA-SE Hereditary Tumors Research Group, Eotvos Lorand Research Network, H-1089 Budapest, Hungary; (B.S.); (I.L.); (H.B.)
- Bionics Innovation Center, H-1089 Budapest, Hungary;
- Department of Laboratory Medicine, Semmelweis University, H-1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, H-1122 Budapest, Hungary
| |
Collapse
|
46
|
Redlich A, Pamporaki C, Lessel L, Frühwald MC, Vorwerk P, Kuhlen M. Pseudohypoxic pheochromocytomas and paragangliomas dominate in children. Pediatr Blood Cancer 2021; 68:e28981. [PMID: 33682326 DOI: 10.1002/pbc.28981] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors that are associated with cancer predisposition syndromes in up to 80% of affected children. PPGLs can be divided into molecularly defined groups with comparable pathogenesis and biology: (1) pseudohypoxic, (2) kinase signaling, and (3) Wnt-altered. METHODS We report the data of children and adolescents diagnosed with PPGL who have been registered with the German GPOH-MET registry since 1997. RESULTS By December 2019, a total of 88 patients with PPGL were reported. Pheochromocytoma occurred in 56%, paraganglioma in 35%, and synchronous PPGLs in 9.1%. A total of 16% of patients presented with lymph node (5.7%) and distant metastases (10%). Median follow-up was 4.2 years (range 0-17.1). Overall and disease-free survival (DFS) were 98.6% and 54.0%, respectively. Local relapses, metastases, and subsequent PPGLs occurred in 11%, 4.5%, and 15% of patients. Germline mutations were detected in 83% of patients (51% in VHL, 21% in SDHB, 7.8% in SDHD, and one patient each in RET and NF1). One patient was diagnosed with Pacak-Zhuang syndrome. A total of 96% of patients presented with PPGL of the pseudohypoxic subgroup (34% TCA cycle-related, 66% VHL/EPAS1-related). In multivariate analyses, extent of tumor resection was a significant prognostic factor for DFS. CONCLUSIONS Most pediatric PPGLs belong to the pseudohypoxia subgroup, which is associated with a high risk of subsequent PPGL events and metastatic disease. Comprehensive molecular profiling of children and adolescents with newly diagnosed PPGLs will open new avenues for personalized diagnosis, treatment, and surveillance.
Collapse
Affiliation(s)
- Antje Redlich
- Pediatric Oncology Department, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Christina Pamporaki
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lienhard Lessel
- Pediatric Oncology Department, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Michael C Frühwald
- Paediatric and Adolescent Medicine, University Medical Center, Augsburg, Germany
| | - Peter Vorwerk
- Pediatric Oncology Department, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Michaela Kuhlen
- Paediatric and Adolescent Medicine, University Medical Center, Augsburg, Germany
| |
Collapse
|
47
|
Lam-Chung CE, Rodríguez LL, Vázquez JA, Chávarri-Guerra Y, Arízaga-Ramírez R, Antonio OF, De Anda González J, López-Hernández MA, Weitzel JN, Castillo D, Gómez-Pérez FJ, Cuevas-Ramos D. A Novel, Likely Pathogenic MAX Germline Variant in a Patient With Unilateral Pheochromocytoma. J Endocr Soc 2021; 5:bvab085. [PMID: 34169220 PMCID: PMC8218934 DOI: 10.1210/jendso/bvab085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Context Inherited MYC-associated factor X (MAX) gene pathogenic variants (PVs) increase risk for pheochromocytomas (PCCs) and/or paragangliomas (PGLs) in adults and children. There is little clinical experience with such mutations. Objective This report highlights an important approach. Methods Clinical assessment, including blood chemistry, imaging studies, and genetic testing were performed. Results A 38-year-old Hispanic woman was diagnosed with PCC in 2015, treated with adrenalectomy, and referred to endocrinology clinic. Notably, she presented to her primary care physician 3 years earlier complaining of left flank pain, intermittent diaphoresis, and holocranial severe headache. We confirmed severe hypertension (180/100 mm Hg) over multiple antihypertensive regimens. Biochemical and radiological studies workup revealed high plasma metanephrine of 255 pg/mL (normal range, < 65 pg/mL) and plasma normetanephrine of 240 pg/mL (normal range, < 196 pg/mL). A noncontrast computed tomography scan of the abdomen revealed a 4.2 × 4.3 × 4.9-cm, round-shaped and heterogenous contrast enhancement of the left adrenal gland, and a 2-mm nonobstructive left kidney stone. A presumptive diagnosis of secondary hypertension was made. After pharmacological therapy, laparoscopic left adrenalectomy was performed and confirmed the diagnosis of pheochromocytoma. Based on her age, family history, and a high suspicion for genetic etiology, genetic testing was performed that revealed the presence of a novel likely pathogenic variant involving a splice consensus sequence in the MAX gene, designated c0.64-2A > G. Conclusion The phenotype of MAX PV-related disease and paraganglioma are highlighted. The novel c0.64-2A > G mutation is reported here and should be considered in the diagnostic workup of similar cases.
Collapse
Affiliation(s)
- César Ernesto Lam-Chung
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Larissa López Rodríguez
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Jazmín Arteaga Vázquez
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Yanin Chávarri-Guerra
- Department of Hemato-Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Rebeca Arízaga-Ramírez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico.,Department of Radiology and Imaging, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Orlando Falcon Antonio
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Jazmín De Anda González
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - María Aurelia López-Hernández
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | | | | | - Francisco Javier Gómez-Pérez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
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
|