1
|
Catalano R, Nozza E, Altieri B, Esposito E, Croci GA, Barbieri AM, Treppiedi D, Di Bari S, Kimpel O, Detomas M, Tamburello M, Schauer MP, Herterich S, Angelousi A, Luconi M, Canu L, Nesi G, Hantel C, Sigala S, Landwehr LS, Di Dalmazi G, Cassinotti E, Baldari L, Palmieri S, Mangone A, Ferrante E, Ronchi CL, Mantovani G, Peverelli E. Emerging role of IGF1R and IR expression and localisation in adrenocortical carcinomas. Cell Commun Signal 2025; 23:119. [PMID: 40038716 PMCID: PMC11877998 DOI: 10.1186/s12964-025-02115-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The insulin-like growth factor 2 (IGF2) is overexpressed in 90% of adrenocortical carcinomas (ACC) and promotes cell proliferation via IGF1R and isoform A of insulin receptor (IRA). However, IGF2 role in ACC tumourigenesis has not been completely understood yet, and the contribution of IGF1R and IRA in mediating ACC cell growth has been poorly explored. This study aimed to investigate IGF1R and IR expression and localisation, including the expression of IR isoforms, in ACC and adrenocortical adenomas (ACA), and their role in IGF2-driven proliferation. METHODS Immunohistochemistry staining of IGF1R and IR was performed on 118 ACC and 22 ACA to evaluate their expression and cellular localisation and statistical analyses were carried out to assess correlations with clinicopathological data. The expression of IRA and IRB in ACC and ACA tissues, ACC cell lines and ACC and ACA primary cultures was determined by RT-qPCR. To appraise the specific role of IGF1R and IR in mediating IGF2 mitogenic pathway, single and double silencing of receptors and their inhibition in 2 ACC cell lines derived from primary tumours (H295R and JIL-2266) and 2 derived from metastatic tumours (MUC-1 and TVBF-7) as well as in ACC and ACA primary cultures were performed. RESULTS We found a higher IGF1R plasma membrane localisation in ACC compared to ACA. In ACC this localisation was associated with higher Ki67 and Weiss score. IR was expressed in about half of ACC and in all ACA but, in ACC, it was associated with higher Ki67 and Weiss score. RT-qPCR revealed that the prevalent isoform of IR was IRA in ACC and ACA, but not in normal adrenals. In ACC cell lines, double IGF1R + IR silencing reduced cell proliferation in JIL-2266, MUC-1 and TVBF-7 but not in H295R. In ACC, but not ACA, primary cultures, cell proliferation was reduced after IR but not IGF1R knockdown. CONCLUSIONS Overall, these data suggest that IGF1R localisation and IR expression represent new biomarkers predicting tumour aggressiveness, as well as possible molecular markers useful to patients' stratification for more individualized IGF1R-IR targeted therapies or for novel pharmacological approaches specifically targeting IRA isoform.
Collapse
Affiliation(s)
- Rosa Catalano
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Emma Nozza
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
- PhD Program in Experimental Medicine, University of Milan, 20122, Milan, Italy
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Emanuela Esposito
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
- PhD Program in Experimental Medicine, University of Milan, 20122, Milan, Italy
| | - Giorgio A Croci
- Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Anna Maria Barbieri
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Donatella Treppiedi
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Sonia Di Bari
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
- PhD Program in Experimental Medicine, University of Milan, 20122, Milan, Italy
| | - Otilia Kimpel
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Mario Detomas
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Mariangela Tamburello
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25121, Brescia, Italy
| | - Marc P Schauer
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Sabine Herterich
- Central Laboratory, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Anna Angelousi
- First Department of Internal Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
- 51st Department of Propaedeutic Internal Medicine, National Technical University of Athens, Mikras Asias 75, Athens, 11527, Greece
| | - Michaela Luconi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50139, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy
| | - Letizia Canu
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50139, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy
| | - Gabriella Nesi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50139, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliero-Universitaria Careggi, 50134, Florence, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, CH-8006, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25121, Brescia, Italy
| | - Laura-Sophie Landwehr
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Guido Di Dalmazi
- Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126, Bologna, Italy
| | - Elisa Cassinotti
- Department of Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Ludovica Baldari
- Department of Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Serena Palmieri
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Alessandra Mangone
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Emanuele Ferrante
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Cristina L Ronchi
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
- Department of Metabolism and System Science, University of Birmingham, Birmingham, B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, B15 2TT, UK
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy.
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy.
| | - Erika Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy.
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy.
| |
Collapse
|
2
|
Jeyaraman K, Concolino P, Falhammar H. Adrenocortical tumors and hereditary syndromes. Expert Rev Endocrinol Metab 2025; 20:1-19. [PMID: 39570085 DOI: 10.1080/17446651.2024.2431748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024]
Abstract
INTRODUCTION Adrenocortical tumors (ACTs) are frequently encountered in clinical practice. They vary in clinical and biological characteristics from nonfunctional to life threatening hormone excess, from benign to highly aggressive malignant tumors. Most ACTs appear to be benign and nonfunctioning. It has been controversial how these apparently benign and nonfunctioning tumors should be monitored. Over the past few decades, significant advances have been made in understanding the regulation of growth and tumorigenesis in adrenocortical cells. Defining the molecular pathomechanisms in inherited tumor syndromes led to the expansion of research to sporadic ACTs. Distinct molecular signatures have been identified in sporadic ACTs and a potential genomic classification of ACT has been proposed. AREAS COVERED In this review, we discuss the various adrenocortical pathologies associated with hereditary syndromes with special focus on their molecular pathomechanisms, the understanding of which is important in the era of precision medicine. EXPERT OPINION Identifying the molecular pathomechanisms of the adrenocortical tumorigenesis in inherited syndromes has led to the understanding of the alterations in different signaling pathways that help explain the wide variations in the biology and behavior of ACTs.
Collapse
Affiliation(s)
| | - Paola Concolino
- Dipartimento di Scienze di Laboratorio ed Ematologiche, UOC Chimica, Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Teng T, Ren L, Xiao J, Shi Z, Li L, Song C. Acute myeloid leukemia cells adhere to bone marrow and acquire chemoresistance by downregulating UNC5B expression. Front Oncol 2024; 14:1394443. [PMID: 39381040 PMCID: PMC11460579 DOI: 10.3389/fonc.2024.1394443] [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: 03/01/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
Acute myeloid leukemia (AML) is a malignant tumor of the hematological system. Because of its characteristics of recurrence, refractory and chemoresistance, new therapeutic targets need to be identified. Adhesion and proliferation are characteristics of AML cells, and critical steps in inducing chemotherapy resistance. In this study, we reported that UNC5B inhibits AML cell bone marrow adhesion, inhibits AML cell proliferation and increases sensitivity to chemotherapy. Mechanistically, RNA sequencing (RNA-seq) and experimental results revealed that overexpression of UNC5B inhibits adhesion and proliferation signaling pathways and inhibits the expression of MPZL1, CLDN23, IGF2 and WNT7B. In conclusion, our findings suggest that UNC5B serves as a prognostic indicator and a potential therapeutic target for AML.
Collapse
Affiliation(s)
- Teng Teng
- Shandong Key Laboratory of Traditional Chinese Medicine and Stress Injury, Department of Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Liping Ren
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jilong Xiao
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyu Shi
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lanbo Li
- Laboratory Animal Center, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Chunhong Song
- Shandong Key Laboratory of Traditional Chinese Medicine and Stress Injury, Department of Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
4
|
Tourigny DS, Altieri B, Secener KA, Sbiera S, Schauer MP, Arampatzi P, Herterich S, Sauer S, Fassnacht M, Ronchi CL. Cellular landscape of adrenocortical carcinoma at single-nuclei resolution. Mol Cell Endocrinol 2024; 590:112272. [PMID: 38759836 DOI: 10.1016/j.mce.2024.112272] [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: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare yet devastating tumour of the adrenal gland with a molecular pathology that remains incompletely understood. To gain novel insights into the cellular landscape of ACC, we generated single-nuclei RNA sequencing (snRNA-seq) data sets from twelve ACC tumour samples and analysed these alongside snRNA-seq data sets from normal adrenal glands (NAGs). We find the ACC tumour microenvironment to be relatively devoid of immune cells compared to NAG tissues, consistent with known high tumour purity values for ACC as an immunologically "cold" tumour. Our analysis identifies three separate groups of ACC samples that are characterised by different relative compositions of adrenocortical cell types. These include cell populations that are specifically enriched in the most clinically aggressive and hormonally active tumours, displaying hallmarks of reorganised cell mechanobiology and dysregulated steroidogenesis, respectively. We also identified and validated a population of mitotically active adrenocortical cells that strongly overexpress genes POLQ, DIAPH3 and EZH2 to support tumour expansion alongside an LGR4+ progenitor-like or cell-of-origin candidate for adrenocortical carcinogenesis. Trajectory inference suggests the fate adopted by malignant adrenocortical cells upon differentiation is associated with the copy number or allelic balance state of the imprinted DLK1/MEG3 genomic locus, which we verified by assessing bulk tumour DNA methylation status. In conclusion, our results therefore provide new insights into the clinical and cellular heterogeneity of ACC, revealing how genetic perturbations to healthy adrenocortical renewal and zonation provide a molecular basis for disease pathogenesis.
Collapse
Affiliation(s)
- David S Tourigny
- School of Mathematics, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Kerim A Secener
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany; Institute of Biochemistry, Department of Biology, Chemistry and Pharmacy, Free University Berlin, Berlin, 14195, Germany
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Marc P Schauer
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany; Center for Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, 97080, Germany
| | | | - Sabine Herterich
- Central Laboratory, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Cristina L Ronchi
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, B15 2GW, UK.
| |
Collapse
|
5
|
Chen G, Li S, Lu J, Liang A, Gao P, Ou F, Wang Y, Li Y, Pan B. LncRNA ZFHX4-AS1 as a novel biomarker in adrenocortical carcinoma. Transl Androl Urol 2024; 13:1188-1205. [PMID: 39100837 PMCID: PMC11291411 DOI: 10.21037/tau-23-649] [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: 12/18/2023] [Accepted: 04/28/2024] [Indexed: 08/06/2024] Open
Abstract
Background Adrenocortical carcinoma (ACC) is a rare and highly aggressive malignant tumor. Currently, there is a lack of reliable prognostic markers in clinical practice. Extensive research has shown that long non-coding RNA (lncRNA) are critical factors in the initiation and progression of cancer, closely associated with early diagnosis and prognosis. Previous studies have identified that ZFHX4 antisense RNA 1 (ZFHX4-AS1) is aberrantly expressed in various cancers and is associated with poor outcomes. This study investigates whether ZFHX4-AS1 affects the prognosis of ACC patients and, if so, the potential mechanisms involved. Methods In this study, utilizing four multi-center cohorts from The Cancer Genome Atlas (TCGA) program and Gene Expression Omnibus (GEO), we validated the prognostic capability of ZFHX4-AS1 in ACC patients through Kaplan-Meier survival analysis, cox regression models, and nomograms. Then, we explored the biological functions of ZFHX4-AS1 using gene set enrichment analysis (GSEA), competing endogenous RNA (ceRNA) networks, and analyses of somatic mutations and copy number variation (CNV). Finally, in vitro experiments were conducted to further validate the impact of ZFHX4-AS1 on proliferation and migration capabilities of ACC cell lines. Results Survival analysis indicated that patients in the high ZFHX4-AS1 expression group of ACC had worse prognosis. Cox regression analyses suggested that ZFHX4-AS1 levels were independent risk factors for prognosis. Subsequently, we constructed nomograms based on clinical features and ZFHX4-AS1 levels, demonstrating good predictive performance under the time-dependent receiver operating characteristic (ROC) curve. Analysis based on somatic mutations and CNV revealed that CTNNB1 and 9p21.3-Del drove the expression of ZFHX4-AS1. Cell Counting Kit-8 (CCK-8), colony formation, and Transwell assays confirmed that knockdown of ZFHX4-AS1 inhibited proliferation and migration of ACC cells. Conclusions This study demonstrates that ZFHX4-AS1 has a reliable predictive value for the prognosis of ACC patients and is a promising biomarker.
Collapse
Affiliation(s)
- Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Songbo Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Anyun Liang
- Department of Andrology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ping Gao
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fengmeng Ou
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yu Wang
- Department of Endocrinology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yutong Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Pan
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
6
|
Tai Y, Shang J. Wnt/β-catenin signaling pathway in the tumor progression of adrenocortical carcinoma. Front Endocrinol (Lausanne) 2024; 14:1260701. [PMID: 38269250 PMCID: PMC10806569 DOI: 10.3389/fendo.2023.1260701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
Adrenocortical carcinoma (ACC) is an uncommon, aggressive endocrine malignancy with a high rate of recurrence, a poor prognosis, and a propensity for metastasis. Currently, only mitotane has received certification from both the US Food and Drug Administration (FDA) and the European Medicines Agency for the therapy of advanced ACC. However, treatment in the advanced periods of the disorders is ineffective and has serious adverse consequences. Completely surgical excision is the only cure but has failed to effectively improve the survival of advanced patients. The aberrantly activated Wnt/β-catenin pathway is one of the catalysts for adrenocortical carcinogenesis. Research has concentrated on identifying methods that can prevent the stimulation of the Wnt/β-catenin pathway and are safe and advantageous for patients in view of the absence of effective treatments and the frequent alteration of the Wnt/β-catenin pathway in ACC. Comprehending the complex connection between the development of ACC and Wnt/β-catenin signaling is essential for accurate pharmacological targets. In this review, we summarize the potential targets between adrenocortical carcinoma and the Wnt/β-catenin signaling pathway. We analyze the relevant targets of drugs or inhibitors that act on the Wnt pathway. Finally, we provide new insights into how drugs or inhibitors may improve the treatment of ACC.
Collapse
Affiliation(s)
- Yanghao Tai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Jiwen Shang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- Department of Ambulatory Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
7
|
Ghosh C, Hu J, Kebebew E. Advances in translational research of the rare cancer type adrenocortical carcinoma. Nat Rev Cancer 2023; 23:805-824. [PMID: 37857840 DOI: 10.1038/s41568-023-00623-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
Adrenocortical carcinoma is a rare malignancy with an annual worldwide incidence of 1-2 cases per 1 million and a 5-year survival rate of <60%. Although adrenocortical carcinoma is rare, such rare cancers account for approximately one third of patients diagnosed with cancer annually. In the past decade, there have been considerable advances in understanding the molecular basis of adrenocortical carcinoma. The genetic events associated with adrenocortical carcinoma in adults are distinct from those of paediatric cases, which are often associated with germline or somatic TP53 mutations and have a better prognosis. In adult primary adrenocortical carcinoma, the main somatic genetic alterations occur in genes that encode proteins involved in the WNT-β-catenin pathway, cell cycle and p53 apoptosis pathway, chromatin remodelling and telomere maintenance pathway, cAMP-protein kinase A (PKA) pathway or DNA transcription and RNA translation pathways. Recently, integrated molecular studies of adrenocortical carcinomas, which have characterized somatic mutations and the methylome as well as gene and microRNA expression profiles, have led to a molecular classification of these tumours that can predict prognosis and have helped to identify new therapeutic targets. In this Review, we summarize these recent translational research advances in adrenocortical carcinoma, which it is hoped could lead to improved patient diagnosis, treatment and outcome.
Collapse
Affiliation(s)
| | - Jiangnan Hu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Electron Kebebew
- Department of Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Abstract
Adrenal cortical carcinoma (ACC) is a rare and aggressive malignancy that poses challenging issues regarding the diagnostic workup. Indeed, no presurgical technique or clinical parameters can reliably distinguish between adrenal cortical adenomas, which are more frequent and have a favorable outcome, and ACC, and the final diagnosis largely relies on histopathologic analysis of the surgical specimen. However, even the pathologic assessment of malignancy in an adrenal cortical lesion is not straightforward and requires a combined evaluation of multiple histopathologic features. Starting from the Weiss score, which was developed in 1984, several histopathologic scoring systems have been designed to tackle the difficulties of ACC diagnosis. Dealing with specific histopathologic variants (eg, Liss-Weiss-Bisceglia scoring system for oncocytic ACC) or patient characteristics (eg, Wieneke index in the pediatric setting), these scores remarkably improved the diagnostic workup of ACC and its subtypes. Nevertheless, cases with misleading features or discordant correlations between pathologic findings and clinical behavior still occur. Owing to multicentric collaborative studies integrating morphologic features with ancillary immunohistochemical markers and molecular analysis, ACC has eventually emerged as a multifaceted, heterogenous malignancy, and, while innovative and promising approaches are currently being tested, the future clinical management of patients with ACC will mainly rely on personalized medicine and target-therapy protocols. At the dawn of the new Fifth World Health Organization classification of endocrine tumors, this review will tackle ACC from the pathologist's perspective, thus focusing on the main available diagnostic, prognostic, and predictive tissue-tethered features and biomarkers and providing relevant clinical and molecular correlates.
Collapse
|
9
|
Altieri B, Lalli E, Faggiano A. Mitotane treatment in adrenocortical carcinoma: mechanisms of action and predictive markers of response to therapy. Minerva Endocrinol (Torino) 2022; 47:203-214. [PMID: 34881855 DOI: 10.23736/s2724-6507.21.03601-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a high risk of recurrence even in cases with complete surgical tumor resection. Mitotane represents the cornerstone of the adjuvant therapy as well as the first line of medical treatment in advanced cases. However, evidence on mitotane efficacy is mostly based on retrospective studies and the use of mitotane continues to represent a clinical challenge. Mitotane causes selective damage to adrenocortical cells, causing an increase of cell apoptosis through a disruption of mitochondria and the induction of the endoplasmic reticulum stress. Different clinical and molecular markers predicting response to mitotane have been proposed with uncertain results. Attainment of mitotane plasma levels within the target range of 14 to 20 mg/L represent the strongest predictor of mitotane effectiveness both in adjuvant and advanced tumor setting. The occurrence of late recurrence after primary ACC diagnosis and changes in metabolic activity on FDG-PET are only weakly associated with mitotane response. Among the proposed molecular markers associated with mitotane efficacy, the investigation of the CYP2W1*6 and CYP2B6*6 single nucleotide polymorphisms appears to be currently the most promising predictive molecular markers of mitotane therapy. However, none of the evaluated markers has been validated for clinical use. In the era of precision medicine, a better insight into mitotane molecular mechanisms as well as the potential use in the daily clinical practice of clinical parameters and molecular markers predicting the individual response to mitotane are urgently needed.
Collapse
Affiliation(s)
- Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany -
| | - Enzo Lalli
- Institute of Molecular and Cellular Pharmacology CNRS UMR, Valbonne, France
- University of Côte d'Azur, Valbonne, France
- INSERM, Valbonne, France
| | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
10
|
Ilanchezhian M, Varghese DG, Glod JW, Reilly KM, Widemann BC, Pommier Y, Kaplan RN, Del Rivero J. Pediatric adrenocortical carcinoma. Front Endocrinol (Lausanne) 2022; 13:961650. [PMID: 36387865 PMCID: PMC9659577 DOI: 10.3389/fendo.2022.961650] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy of the adrenal gland with an unfavorable prognosis. It is rare in the pediatric population, with an incidence of 0.2-0.3 patients per million in patients under 20 years old. It is primarily associated with Li-Fraumeni and Beckwith-Wiedemann tumor predisposition syndromes in children. The incidence of pediatric ACC is 10-15fold higher in southern Brazil due to a higher prevalence of TP53 mutation associated with Li-Fraumeni syndrome in that population. Current treatment protocols are derived from adult ACC and consist of surgery and/or chemotherapy with etoposide, doxorubicin, and cisplatin (EDP) with mitotane. Limited research has been reported on other treatment modalities for pediatric ACC, including mitotane, pembrolizumab, cabozantinib, and chimeric antigen receptor autologous cell (CAR-T) therapy.
Collapse
Affiliation(s)
- Maran Ilanchezhian
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Diana Grace Varghese
- Developmental Therapeutics Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - John W. Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Karlyne M. Reilly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Yves Pommier
- Developmental Therapeutics Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Rosandra N. Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
- *Correspondence: Jaydira Del Rivero,
| |
Collapse
|
11
|
The Challenging Pharmacokinetics of Mitotane: An Old Drug in Need of New Packaging. Eur J Drug Metab Pharmacokinet 2021; 46:575-593. [PMID: 34287806 PMCID: PMC8397669 DOI: 10.1007/s13318-021-00700-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 01/10/2023]
Abstract
Adrenocortical carcinoma (ACC) is a malignant tumor originating from the adrenal gland cortex with a heterogeneous but overall dismal prognosis in advanced stages. For more than 50 years, mitotane has remained a cornerstone for the treatment of ACC as adjuvant and palliative therapy. It has a very poor aqueous solubility of 0.1 mg/l and high partition coefficient in octanol/water (log P) value of 6. The commercially available dosage form is 500 mg tablets (Lysodren®). Even at doses up to 6 g/day (12 tablets in divided doses) for several months, > 50% patients do not achieve therapeutic plasma concentration > 14 mg/l due to poor water solubility, large volume of distribution and inter/intra-individual variability in bioavailability. This article aims to give a concise update of the clinical challenges associated with the administration of high-dose mitotane oral therapy which encompass the issues of poor bioavailability, difficult-to-predict pharmacokinetics and associated adverse events. Moreover, we present recent efforts to improve mitotane formulations. Their success has been limited, and we therefore propose an injectable mitotane formulation instead of oral administration, which could bypass many of the main issues associated with high-dose oral mitotane therapy. A parenteral administration of mitotane could not only help to alleviate the adverse effects but also circumvent the variable oral absorption, give better control over therapeutic plasma mitotane concentration and potentially shorten the time to achieve therapeutic drug plasma concentrations considerably. Mitotane as tablet form is currently the standard treatment for adrenocortical carcinoma. It has been used for 5 decades but suffers from highly variable responses in patients, subsequent adverse effects and overall lower response rate. This can be fundamentally linked to the exceedingly poor water solubility of mitotane itself. In terms of enhancing water solubility, a few research groups have attempted to develop better formulations of mitotane to overcome the issues associated with tablet dosage form. However, the success rate was limited, and these formulations did not make it into the clinics. In this article, we have comprehensively reviewed the properties of these formulations and discuss the reasons for their limited utility. Furthermore, we discuss a recently developed mitotane nanoformulation that led us to propose a novel approach to mitotane therapy, where intravenous delivery supplements the standard oral administration. With this article, we combine the current state of knowledge as a single piece of information about the various problems associated with the use of mitotane tablets, and herein we postulate the development of a new injectable mitotane formulation, which can potentially circumvent the major problems associated to mitotane's poor water solubility.
Collapse
|
12
|
Oliveira RC, Martins MJ, Moreno C, Almeida R, Carvalho J, Teixeira P, Teixeira M, Silva ET, Paiva I, Figueiredo A, Cipriano MA. Histological scores and tumor size on stage II in adrenocortical carcinomas. Rare Tumors 2021; 13:20363613211026494. [PMID: 34262677 PMCID: PMC8243092 DOI: 10.1177/20363613211026494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/01/2021] [Indexed: 11/18/2022] Open
Abstract
Adrenocortical carcinomas (ACC) are aggressive tumors with a poor prognosis.
Histological scores are advised for the diagnosis, however, there are borderline
cases that may be misjudged as adrenocortical adenomas (ACA). The three main
scores used are: Weiss Modified System (WMS), Reticulin Algorithm (RA), and
Helsinki Score (HS). We intend to compare the accuracy of the three scores in
ACC diagnosis and to identify predictive factors of overall survival (OS).
Retrospective study (2004–2016) at Centro Hospitalar e Universitário de Coimbra
of the adrenal tumors, classified as ACC or ACA, with a history of posterior
tumor relapse/metastases, without lesions in the contralateral adrenal gland:
13F and 6M, with a median age of 51 ± 12.41 years. Nodules’ median size was
9.20 ± 6.2 cm. Patients had a median OS of 52 ± 18.6 months, with 57.9% and
46.3%, at 3 and 5 years. Seven patients had local recurrence and nine had
metastases. Thirteen cases were in stage II. The WMS and the HS allowed a
diagnosis of ACC in 15 cases and the RA defined ACC in 17 cases. All cases had,
at least, focal disruption of the reticulin framework. More than
5 mitosis/50 HPF was associated with worse OS: 49.67 ± 21.43 versus
108.86 ± 14.02 months (p = 0.026). In patients with stage II,
tumor size ⩾10 cm was associated with worse OS: 19.25 ± 7.15 versus
96.11 ± 16.7 months (p = 0.007), confirmed by multivariate
analysis (p = 0.031). The correct diagnosis of ACC is a
pathologist responsibility. The RA seems the most accurate. Any loss of the
reticulin framework should raise awareness for malignancy. In patients on stage
II, a size ⩾10 cm is a predictor of worse prognosis.
Collapse
Affiliation(s)
- Rui Caetano Oliveira
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria João Martins
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - Carolina Moreno
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal.,Endocrinology, Diabetes and Metabolism Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Rui Almeida
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - João Carvalho
- Urology and Renal Transplantation, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Paulo Teixeira
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Teixeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Edgar Tavares Silva
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal.,Urology and Renal Transplantation, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Isabel Paiva
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - Arnaldo Figueiredo
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal.,Urology and Renal Transplantation, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | |
Collapse
|
13
|
Min SK, Kim M, Park JB. Insulin-like growth factor 2-enhanced osteogenic differentiation of stem cell spheroids by regulation of Runx2 and Col1 expression. Exp Ther Med 2021; 21:383. [PMID: 33680105 PMCID: PMC7918416 DOI: 10.3892/etm.2021.9814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor 2 (IGF-2) is a growth factor that is involved in various functions of cells, including stem cells. The effects of IGF-2 on the cellular viability and osteogenic differentiation of stem cell spheroids were investigated in the present study. Stem cell spheroids were formed using concave microwells in the presence of IGF-2 at final concentrations of 0, 10 and 100 ng/ml. Cellular viability was measured qualitatively using a microscope and quantitatively using an assay kit based on water-soluble tetrazolium salt. The level of alkaline phosphatase activity, and an anthraquinone dye assay for calcium deposit evaluation, were used to assess osteogenic differentiation. A quantitative PCR analysis was conducted to evaluate the expression of Runx2 and Col1. Spheroid formation was noticed on day 1 in the microwells, and the spheroidal shape was maintained up to day 7. The cell viability assay values for IGF-2 at 0, 10 and 100 ng/ml at day 1 were 0.193±0.002, 0.191±0.002 and 0.201±0.006, respectively (P>0.05). The absorbance values at 405 nm for the alkaline phosphatase activity assays on day 21 were 0.221±0.006, 0.375±0.010 and 0.280±0.015 for IGF-2 at 0, 10 and 100 ng/ml, respectively. There were significantly higher values for IGF-2 in the 10 and 100 ng/ml groups when compared with the control (P<0.05). Significantly higher Alizarin red staining was noted for IGF-2 in the 10 ng/ml group when compared with the unloaded control at day 21 (P<0.05). Quantitative PCR revealed that mRNA levels of Runx2 and Col1 were significantly higher at 100 ng/ml on day 7. Conclusively, the present study demonstrated that the application of IGF-2 increased alkaline phosphatase activity, Alizarin red staining, and Runx2 and Col1 expression of stem cell spheroids.
Collapse
Affiliation(s)
- Sae Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Minji Kim
- College of Dentistry, Chosun University, Gwangju 61452, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
14
|
Mizdrak M, Tičinović Kurir T, Božić J. The Role of Biomarkers in Adrenocortical Carcinoma: A Review of Current Evidence and Future Perspectives. Biomedicines 2021; 9:174. [PMID: 33578890 PMCID: PMC7916711 DOI: 10.3390/biomedicines9020174] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy arising from the adrenal cortex often with unexpected biological behavior. It can occur at any age, with two peaks of incidence: in the first and between fifth and seventh decades of life. Although ACC are mostly hormonally active, precursors and metabolites, rather than end products of steroidogenesis are produced by dedifferentiated and immature malignant cells. Distinguishing the etiology of adrenal mass, between benign adenomas, which are quite frequent in general population, and malignant carcinomas with dismal prognosis is often unfeasible. Even after pathohistological analysis, diagnosis of adrenocortical carcinomas is not always straightforward and represents a great challenge for experienced and multidisciplinary expert teams. No single imaging method, hormonal work-up or immunohistochemical labelling can definitively prove the diagnosis of ACC. Over several decades' great efforts have been made in finding novel reliable and available diagnostic and prognostic factors including steroid metabolome profiling or target gene identification. Despite these achievements, the 5-year mortality rate still accounts for approximately 75% to 90%, ACC is frequently diagnosed in advanced stages and therapeutic options are unfortunately limited. Therefore, imperative is to identify new biological markers that can predict patient prognosis and provide new therapeutic options.
Collapse
Affiliation(s)
- Maja Mizdrak
- Department of Nephrology and Hemodialysis, University Hospital of Split, 21000 Split, Croatia;
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| | - Tina Tičinović Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
- Department of Endocrinology, Diabetes and Metabolic Disorders, University Hospital of Split, 21000 Split, Croatia
| | - Joško Božić
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia;
| |
Collapse
|
15
|
Lazúrová I, Jochmanová I, Sotak Š, Špaková I, Mareková M. Is there a role for the IGF system and epidermal growth factor (EGF) in the pathogenesis of adrenocortical adenomas? A preliminary case-control study. Physiol Res 2020; 69:1085-1094. [PMID: 33210933 PMCID: PMC8549875 DOI: 10.33549/physiolres.934553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/29/2020] [Indexed: 11/25/2022] Open
Abstract
Adrenal incidentalomas (AI) are very common and mostly they are non-functioning adenomas (NFA). NFAs are often associated with insulin resistance and metabolic syndrome. Several biomarkers, including certain growth factors, may participatein the pathogenesis ofmetabolic changes in patients with adrenal adenomas.Patients with NFA and age-matched control subjects were enrolled in the study. Data on age, gender, presence of metabolic syndrome or its components were obtained for each subject. Blood samples were obtained and glycemia, insulinemia, lipid profile, and selected growth factor levels were measured. Forty-three patients with NFA and 40 controls were included in the study. Differences were not found in the metabolic syndrome and its components prevalence or in the biochemical profile between patients and the control group. Significant differences were noticed in the levels of IGF1, IGF2, and IGFBP3 (p=0.016, p=0.005, p=0.004, respectively), but there were no differences in VEGF or EGF concentrations. In NFA patients, an association between glycemia and EGF levels was present (p=0.026). No significant correlations between tumor size and insulin or growth factor concentrations were present in AI patients. Significantly higher serum IGF1, IGF2, and IGFBP3 concentrations in NFA patients may support the role of the IGF axis in the pathogenesis of adrenocortical lesions.No correlation between IGFs or IGFBP3 and parameters of glucose or lipid metabolism was found. Present results may support the role of the growth hormone axis rather than hyperinsulinemia and insulin resistance in the pathogenesis of adrenocortical adenomas.
Collapse
Affiliation(s)
- I Lazúrová
- First Department of Internal Medicine, Faculty of Medicine, P.J. Šafárik University, Košice, Slovakia.
| | | | | | | | | |
Collapse
|
16
|
The cytoskeleton actin binding protein filamin A impairs both IGF2 mitogenic effects and the efficacy of IGF1R inhibitors in adrenocortical cancer cells. Cancer Lett 2020; 497:77-88. [PMID: 33075426 DOI: 10.1016/j.canlet.2020.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
Adrenocortical carcinomas (ACCs) overexpress insulin-like growth factor 2 (IGF2), that drives a proliferative autocrine loop by binding to IGF1R and IR, but IGF1R/IR-targeted therapies failed in ACC patients. The cytoskeleton actin-binding protein filamin A (FLNA) impairs IR signalling in melanoma cells. Aims of this study were to test FLNA involvement in regulating IGF1R and IR responsiveness to both IGF2 and inhibitors in ACC. In ACC cells H295R and SW13 and primary cultures (1ACC, 4 adenomas) we found that IGF1R and IR interacted with FLNA, and FLNA silencing increased IGF1R and reduced IR expression, with a downstream effect of increased cell proliferation and ERK phosphorylation. In addition, FLNA knockdown potentiated antiproliferative effects of IGF1R/IR inhibitor Linsitinib and IGF1R inhibitor NVP-ADW742 in H295R. Finally, Western blot showed lower FLNA expression in ACCs (n = 10) than in ACAs (n = 10) and an inverse correlation of FLNA/IGF1R ratio with ERK phosphorylation in ACCs only. In conclusion, we demonstrated that low FLNA levels enhance both IGF2 proliferative effects and IGF1R/IR inhibitors efficacy in ACC cells, suggesting FLNA as a new factor influencing tumor clinical behavior and the response to the therapy with IGF1R/IR-targeted drugs.
Collapse
|
17
|
Pereira SS, Monteiro MP, Costa MM, Moreira Â, Alves MG, Oliveira PF, Jarak I, Pignatelli D. IGF2 role in adrenocortical carcinoma biology. Endocrine 2019; 66:326-337. [PMID: 31378849 PMCID: PMC6838304 DOI: 10.1007/s12020-019-02033-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/20/2019] [Indexed: 11/24/2022]
Abstract
PURPOSE Clinical outcomes of adrenocortical carcinomas (ACC) could be improved by using novel treatment targets based on the recent advances of tumor biology knowledge. Insulin-like growth factor 2 (IGF2) protein expression is usually 8-80 fold higher in ACC when compared to normal adrenal glands (N-AG) or adrenocortical adenomas (ACA), despite the fact that the biological features of high vs. low IGF2 expressing ACC have not yet been well characterized. Our goal was to understand the IGF2 role in ACC biology by focusing in several cancer hallmarks, including cell proliferation, viability, invasion, and metabolism. METHODS IGF2 immunohistochemistry expression was evaluated in ACC (n = 13), non-functioning adrenocortical adenoma (ACAn) (n = 14), and N-AG (n = 9). The effects of IGF2 (50, 100 ng/mL) in cell proliferation, viability, invasion, and metabolism, as well as in MAPK/ERK and mTOR pathways activation and N-cadherin expression, were evaluated in the ACC human cell line H295R. RESULTS IGF2 expression was increased in ACC compared to ACAn and N-AG. Exposure to 100 ng/mL of IGF2 increased H295R cell proliferation and viability. mTOR inhibition reverted IGF2 triggered cell proliferation and viability while MEK/MAPK/ERK inhibition only reverted IGF2 effects on cell proliferation. IGF2 at a 50 ng/mL concentration increased the glycolytic flux and decreased glutamine consumption. CONCLUSIONS IGF2 is an excellent marker to differentiate ACC from ACAn. In addition, IGF2 was demonstrated to influence adrenocortical cancer cell proliferation, metabolism, and viability, but not the cell invasion. These data support that different IGF2 concentrations in ACC can be responsible for different biological behaviors of ACC.
Collapse
Affiliation(s)
- Sofia S Pereira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), ICBAS, University of Porto, Porto, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), ICBAS, University of Porto, Porto, Portugal
| | - Madalena M Costa
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), ICBAS, University of Porto, Porto, Portugal
| | - Ângela Moreira
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), ICBAS, University of Porto, Porto, Portugal
| | - Marco G Alves
- Biology and Genetics of Reproduction, Department of Microscopy, Laboratory of Cell Biology, Multidisciplinary Unit for Biomedical Research (UMIB), ICBAS, University of Porto, Porto, Portugal
- Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Pedro F Oliveira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
| | - Ivana Jarak
- Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Duarte Pignatelli
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal.
- Department of Endocrinology, Hospital S.João, Porto, Portugal.
| |
Collapse
|
18
|
Abstract
Adrenocortical carcinoma (ACC) is an uncommon and heterogeneous disease and may present differently in children and adults. Management of ACC is dependent on disease stage and complete surgical resection is the only potentially curative treatment. The first and most extensively used adrenocortical cancer cell line, as model system to examine mechanisms controlling normal and pathologic function of adrenal cortex, was initially isolated in 1980. Although NCI-H295 maintained steroid capabilities and adrenocortical characteristics, the lack of new cell lines and animal models of ACC has hampered the progress and development of new therapies. In this review we provide description of cellular and patient-derived tumor xenograft (PDTX) models of ACC generated for the elucidation of the underlying pathogenic mechanisms and preclinical functional studies for this aggressive disease.
Collapse
|
19
|
Lotfi CFP, Kremer JL, dos Santos Passaia B, Cavalcante IP. The human adrenal cortex: growth control and disorders. Clinics (Sao Paulo) 2018; 73:e473s. [PMID: 30208164 PMCID: PMC6113920 DOI: 10.6061/clinics/2018/e473s] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
This review summarizes key knowledge regarding the development, growth, and growth disorders of the adrenal cortex from a molecular perspective. The adrenal gland consists of two distinct regions: the cortex and the medulla. During embryological development and transition to the adult adrenal gland, the adrenal cortex acquires three different structural and functional zones. Significant progress has been made in understanding the signaling and molecules involved during adrenal cortex zonation. Equally significant is the knowledge obtained regarding the action of peptide factors involved in the maintenance of zonation of the adrenal cortex, such as peptides derived from proopiomelanocortin processing, adrenocorticotropin and N-terminal proopiomelanocortin. Findings regarding the development, maintenance and growth of the adrenal cortex and the molecular factors involved has improved the scientific understanding of disorders that affect adrenal cortex growth. Hypoplasia, hyperplasia and adrenocortical tumors, including adult and pediatric adrenocortical adenomas and carcinomas, are described together with findings regarding molecular and pathway alterations. Comprehensive genomic analyses of adrenocortical tumors have shown gene expression profiles associated with malignancy as well as methylation alterations and the involvement of miRNAs. These findings provide a new perspective on the diagnosis, therapeutic possibilities and prognosis of adrenocortical disorders.
Collapse
Affiliation(s)
- Claudimara Ferini Pacicco Lotfi
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Jean Lucas Kremer
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Barbara dos Santos Passaia
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Isadora Pontes Cavalcante
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
20
|
Jonker PKC, Meyer VM, Kruijff S. Epigenetic dysregulation in adrenocortical carcinoma, a systematic review of the literature. Mol Cell Endocrinol 2018; 469:77-84. [PMID: 28830787 DOI: 10.1016/j.mce.2017.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/17/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy with a poor prognosis. Diagnosis and treatment of this tumor remains challenging. The Weiss score, the current gold standard for the histopathological diagnosis of ACC, lacks diagnostic accuracy of borderline tumors (Weiss score 2 or 3) and is subject to inter observer variability. Furthermore, adjuvant and palliative systemic therapy have limited effect and no proven overall survival benefit. A better insight in the molecular background of ACC might identify markers that improve diagnostic accuracy, predict treatment response or even provide novel therapeutic targets. This systematic review of the literature aims to provide an overview of alterations in DNA methylation, histone modifications and their potential clinical relevance in ACC.
Collapse
Affiliation(s)
- P K C Jonker
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - V M Meyer
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - S Kruijff
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Altieri B, Colao A, Faggiano A. The role of insulin-like growth factor system in the adrenocortical tumors. MINERVA ENDOCRINOL 2018; 44:43-57. [PMID: 29963827 DOI: 10.23736/s0391-1977.18.02882-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The different presentation of adrenocortical tumors in benign adenoma (ACA) or adrenocortical carcinoma (ACC) is related to the variability at the molecular level. The insulin-like growth factor (IGF) system is one of the most frequently altered pathways in ACC. In this review we will critically analyze the evidence regarding the pathogenic role of the IGF system in adrenal tumorigenesis, focusing on ACC. We will also examine the preclinical and clinical studies which investigated the targeting of the IGF system as a therapeutic approach in ACC. EVIDENCE ACQUISITION The IGF system plays a crucial role in the embryogenesis of adrenal glands. No significant alterations of the IGF system were observed in ACA. In ACC, the IGF2 overexpression is one of the most frequent molecular change presented in more than 85% of cases. However, IGF2 seems to be only a tumor progression factor which requires additional hits to trigger adrenal tumorigenesis. Also, the IGF1 receptor (IGF1R) appears to be higher expressed in ACC. Many IGF1R target-drugs have been developed to inhibit the activation of the IGF system. EVIDENCE SYNTHESIS Preclinical studies using antibody or tyrosine kinase which target the IGF1R, or the dual-targeting of IGF1R and insulin receptor (IR) reduced ACC cells proliferation both in vitro and in vivo in mouse xenograft model. However, these promising results were not confirmed in clinical trials. CONCLUSIONS Nowadays, predictive markers for the response of target-IGF therapy are missing and further studies which investigate new molecular markers and evaluate the entire IGF receptors, including the IR, are urgently needed.
Collapse
Affiliation(s)
- Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Wuerzburg, Wuerzburg, Germany - .,Department of Clinical Medicine and Surgery, University "Federico II", Naples, Italy -
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, University "Federico II", Naples, Italy
| | - Antongiulio Faggiano
- Department of Clinical Medicine and Surgery, University "Federico II", Naples, Italy
| |
Collapse
|
22
|
Komarowska H, Rucinski M, Tyczewska M, Sawicka-Gutaj N, Szyszka M, Hernik A, Klimont A, Milecka P, Migasiuk L, Biczysko M, Idasiak-Piechocka I, Karczewski M, Ruchala M. Ghrelin as a potential molecular marker of adrenal carcinogenesis: In vivo and in vitro evidence. Clin Endocrinol (Oxf) 2018; 89:36-45. [PMID: 29682767 DOI: 10.1111/cen.13725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/30/2022]
Abstract
CONTEXT Adrenal tumours belong to one of the most prevalent neoplasms. It is a heterogeneous group with different aetiology, clinical manifestation and prognosis. Its histopathologic diagnosis is difficult and identification of differentiation markers for tumorigenesis is extremely valuable for diagnosis. DESIGN To assess ghrelin expression and the relationship among ghrelin, IGF2 and the clinicopathological characteristics of adrenal tumours. To investigate the influence of ghrelin on ACC cell line proliferation. MATERIALS AND METHODS Expression of ghrelin and IGF2 in a total of 84 adrenal tissue samples (30 adenoma, 12 hyperplasia, 8 myelolipoma, 20 pheochromocytoma, 7 carcinoma and 7 unchanged adrenal glands) were estimated. Every operated patient from whom samples were obtained underwent clinicopathological analysis. All the parameters were compared among the groups examined and correlations between these were estimated. H295R cell line was incubated with ghrelin to assess its effect on proliferation and migration rate. RESULTS The highest ghrelin expression was observed in carcinoma samples and the lowest in the control group. Ghrelin expression was 21 times higher in carcinoma (P = .017) and 2.4 times higher in adenoma (P = .029) compared with controls. There were no statistically significant differences between myelolipoma (P = .093) and pheochromocytoma (P = .204) relative to the control. Ghrelin level was significantly higher in carcinoma compared to adenoma (P = .049) samples. A positive correlation between ghrelin and IGF2 expression was observed only in myelolipoma (P = .001). Ghrelin at concentrations of 1 × 10-6 mol/L and 1 × 10-8 mol/L significantly stimulated proliferation and migration rate in the H295R cell line. CONCLUSION Ghrelin appears to be an essential factor in driving adrenal tumours development.
Collapse
Affiliation(s)
- Hanna Komarowska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Hernik
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Klimont
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Paulina Milecka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Laura Migasiuk
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Mateusz Biczysko
- Department of General, Endocrinological and Gastroenterological Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Ilona Idasiak-Piechocka
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Karczewski
- Department of General and Transplantation Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchala
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
23
|
Li T, Wang J, Liu P, Chi J, Yan H, Lei L, Li Z, Yang B, Wang X. Insulin-like growth factor 2 axis supports the serum-independent growth of malignant rhabdoid tumor and is activated by microenvironment stress. Oncotarget 2018; 8:47269-47283. [PMID: 28521298 PMCID: PMC5564563 DOI: 10.18632/oncotarget.17617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/18/2017] [Indexed: 11/30/2022] Open
Abstract
Malignant rhabdoid tumors (MRTs) are rare, lethal, pediatric tumors predominantly found in the kidney, brain and soft tissues. MRTs are driven by loss of tumor suppressor SNF5/INI1/SMARCB1/BAF47. The prognosis of MRT is poor using currently available treatments, so new treatment targets need to be identified to expand treatment options for patients experiencing chemotherapy resistance. The growth hormone insulin-like growth factor 2 (IGF2) signaling pathway is a promising target to overcome drug resistance in many cancers. Here, we evaluated the role of IGF2 axis in MRT cell proliferation. We showed that microenvironment stress, including starvation treatment and chemotherapy exposure, lead to elevated expression of IGF2 in the SNF5-deficient MRT cell line. The autocrine IGF2, in turn, activated insulin-like growth factor 1 receptor (IGF1R), insulin receptor (INSR), followed by PI3K/AKT pathway and RAS/ERK pathway to promote cancer cell proliferation and survival. We further demonstrated that impairment of IGF2 signaling by IGF2 neutralizing antibody, IGF1R inhibitor NVP-AEW541 or AKT inhibitor MK-2206 2HCl treatment prevented MRT cell growth in vitro. Taken together, our characterization of this axis defines a novel mechanism for MRT cell growth in the microenvironment of stress. Our results also demonstrated the necessity to test the treatment effect targeting this axis in future research.
Collapse
Affiliation(s)
- Ting Li
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Jin Wang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Pengfei Liu
- Department of Lymphoma, Sino-Us Center of Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiadong Chi
- Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Han Yan
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Lei Lei
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Zexing Li
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Bing Yang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| | - Xi Wang
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Laboratory of Epigenetics in Development and Tumorigenesis, Tianjin Research Center of Basic Medical Sciences, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
24
|
Agosta C, Laugier J, Guyon L, Denis J, Bertherat J, Libé R, Boisson B, Sturm N, Feige JJ, Chabre O, Cherradi N. MiR-483-5p and miR-139-5p promote aggressiveness by targeting N-myc downstream-regulated gene family members in adrenocortical cancer. Int J Cancer 2018. [PMID: 29516499 DOI: 10.1002/ijc.31363] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adrenocortical carcinoma (ACC) is a tumor with poor prognosis in which overexpression of a panel of microRNAs has been associated with malignancy but a very limited number of investigations on their role in ACC pathogenesis have been conducted. We examined the involvement of miR-483-5p and miR-139-5p in adrenocortical cancer aggressiveness. Using bioinformatics predictions and mRNA/miRNA expression profiles, we performed an integrated analysis to identify inversely correlated miRNA-mRNA pairs in ACC. We identified N-myc downstream-regulated gene family members 2 and 4 (NDRG2 and NDRG4) as targets of miR-483-5p and miR-139-5p, respectively. NDRG2 and NDRG4 expressions were inversely correlated respectively with miR-483-5p and miR-139-5p levels in aggressive ACC samples from two independent cohorts of 20 and 44 ACC. Moreover, upregulation of miR-139-5p and downregulation of NDRG4 demonstrated a striking prognostic value. A direct interaction between miR-483-5p or miR-139-5p and their targets was demonstrated in reporter assays. Downregulation of miR-483-5p or miR-139-5p in the ACC cell lines NCI-H295R and SW13 increased NDRG2 or NDRG4 mRNA and protein expression, compromised adrenocortical cancer cell invasiveness and anchorage-independent growth. MiR-483-5p or miR-139-5p overexpression and NDRG2 or NDRG4 inhibition produce similar changes, which are rescued by NDRG2 or NDRG4 ectopic expression. We established that key factors mediating epithelial-to-mesenchymal transition are downstream effectors of miR-483-5p/NDRG2 and miR-139-5p/NDRG4 pathways. Collectively, our data show for the first time that miR-483-5p/NDRG2 and miR-139-5p/NDRG4 axes promote ACC aggressiveness, with potential implications for prognosis and therapeutic interventions in adrenocortical malignancies.
Collapse
Affiliation(s)
- Claire Agosta
- Centre Hospitalier Universitaire Grenoble Alpes, Service d'Endocrinologie, Grenoble, France.,Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Jonathan Laugier
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Laurent Guyon
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Josiane Denis
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Jérôme Bertherat
- Université Paris Descartes, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique UMR 8104, Unité 1016, Institut Cochin, Paris, France.,Département d'Endocrinologie, Centre Expert Cancers Rares de la Surrénale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Rossella Libé
- Université Paris Descartes, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique UMR 8104, Unité 1016, Institut Cochin, Paris, France.,Département d'Endocrinologie, Centre Expert Cancers Rares de la Surrénale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Bruno Boisson
- Centre Hospitalier Universitaire Grenoble Alpes, Institut de Biologie et de Pathologie, Grenoble, France
| | - Nathalie Sturm
- Centre Hospitalier Universitaire Grenoble Alpes, Institut de Biologie et de Pathologie, Grenoble, France
| | - Jean-Jacques Feige
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Olivier Chabre
- Centre Hospitalier Universitaire Grenoble Alpes, Service d'Endocrinologie, Grenoble, France.,Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| | - Nadia Cherradi
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Commissariat à l'Energie Atomique, Biologie du Cancer et de l'Infection, Institut de Biosciences et Biotechnologies de Grenoble, Grenoble, France.,Université Grenoble Alpes, Unité Mixte de Recherche-S1036, Grenoble, France
| |
Collapse
|
25
|
Yak IGF2 Promotes Fibroblast Proliferation Via Suppression of IGF1R and PI3KCG Expression. Genes (Basel) 2018; 9:genes9030169. [PMID: 29558395 PMCID: PMC5867890 DOI: 10.3390/genes9030169] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/03/2018] [Accepted: 03/12/2018] [Indexed: 11/29/2022] Open
Abstract
Insulin-like growth factor 2 (IGF2) recapitulates many of the activities of insulin and promotes differentiation of myoblasts and osteoblasts, which likely contribute to genetic variations of growth potential. However, little is known about the functions and signaling properties of IGF2 variants in yaks. The over-expression vector and knockdown sequence of yak IGF2 were transfected into yak fibroblasts, and the effects were detected by a series of assays. IGF2 expression in yak muscle tissues was significantly lower than that of other tissues. In yak fibroblasts, the up-regulated expression of IGF2 inhibits expression of IGF1 and insulin-like growth factor 2 receptor (IGF2R) and significantly up-regulates expression of IGF1R. Inhibition of IGF2 expression caused the up-regulates expression of IGF1, IGF1R and IGF2R. Both over-expression and knockdown of IGF2 resulted in up-regulation of threonine protein kinase 1 (Akt1) expression and down-regulation of phosphatidylinositol 3-kinase, catalytic subunit gamma (PIK3CG). Cell cycle and cell proliferation assays revealed that over-expression of IGF2 enhanced the DNA synthesis phase and promoted yak fibroblasts proliferation. Conversely, knockdown of IGF2 decreased DNA synthesis and inhibited proliferation. These results suggested that IGF2 was negatively correlated with IGF1R and PIK3CG and demonstrated an association with the IGFs-PI3K-Akt (IGFs-phosphatidylinositol 3-kinase- threonine protein kinase) pathway in cell proliferation and provided evidence supporting the functional role of IGF2 for use in improving the production performance of yaks.
Collapse
|
26
|
Bonnet-Serrano F, Bertherat J. Genetics of tumors of the adrenal cortex. Endocr Relat Cancer 2018; 25:R131-R152. [PMID: 29233839 DOI: 10.1530/erc-17-0361] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/12/2017] [Indexed: 01/23/2023]
Abstract
This review describes the molecular alterations observed in the various types of tumors of the adrenal cortex, excluding Conn adenomas, especially the alterations identified by genomic approaches these last five years. Two main forms of bilateral adrenocortical tumors can be distinguished according to size and aspect of the nodules: primary pigmented nodular adrenal disease (PPNAD), which can be sporadic or part of Carney complex and primary bilateral macro nodular adrenal hyperplasia (PBMAH). The bilateral nature of the tumors suggests the existence of an underlying genetic predisposition. PPNAD and Carney complex are mainly due to germline-inactivating mutations of PRKAR1A, coding for a regulatory subunit of PKA, whereas PBMAH genetic seems more complex. However, genome-wide approaches allowed the identification of a new tumor suppressor gene, ARMC5, whose germline alteration could be responsible for at least 25% of PBMAH cases. Unilateral adrenocortical tumors are more frequent, mostly adenomas. The Wnt/beta-catenin pathway can be activated in both benign and malignant tumors by CTNNB1 mutations and by ZNRF3 inactivation in adrenal cancer (ACC). Some other signaling pathways are more specific of the tumor dignity. Thus, somatic mutations of cAMP/PKA pathway genes, mainly PRKACA, coding for the catalytic alpha-subunit of PKA, are found in cortisol-secreting adenomas, whereas IGF-II overexpression and alterations of p53 signaling pathway are observed in ACC. Genome-wide approaches including transcriptome, SNP, methylome and miRome analysis have identified new genetic and epigenetic alterations and the further clustering of ACC in subgroups associated with different prognosis, allowing the development of new prognosis markers.
Collapse
Affiliation(s)
- Fidéline Bonnet-Serrano
- Institut CochinINSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
- Hormonal Biology LaboratoryAssistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Jérôme Bertherat
- Institut CochinINSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
- Department of EndocrinologyAssistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| |
Collapse
|
27
|
Costa R, Carneiro BA, Tavora F, Pai SG, Kaplan JB, Chae YK, Chandra S, Kopp PA, Giles FJ. The challenge of developmental therapeutics for adrenocortical carcinoma. Oncotarget 2018; 7:46734-46749. [PMID: 27102148 PMCID: PMC5216833 DOI: 10.18632/oncotarget.8774] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/10/2016] [Indexed: 12/11/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare disease with an estimated incidence of only 0.7 new cases per million per year. Approximately 30-70% of the patients present with advanced disease with very poor prognosis and without effective therapeutic options. In the recent years, unprecedented progresses in cancer biology and genomics have fostered the development of numerous targeted therapies for various malignancies. Immunotherapy has also transformed the treatment landscape of malignancies such as melanoma, among others. However, these advances have not brought meaningful benefits for patients with ACC. Extensive genomic analyses of ACC have revealed numerous signal transduction pathway aberrations (e.g., insulin growth factor receptor and Wnt/β-catenin pathways) that play a central role in pathophysiology. These molecular alterations have been explored as potential therapeutic targets for drug development. This manuscript summarizes recent discoveries in ACC biology, reviews the results of early clinical studies with targeted therapies, and provides the rationale for emerging treatment strategies such as immunotherapy.
Collapse
Affiliation(s)
- Ricardo Costa
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Benedito A Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Fabio Tavora
- Department of Pathology, Messejana Heart and Lung Hospital, Fortaleza, Brazil
| | - Sachin G Pai
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jason B Kaplan
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Young Kwang Chae
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sunandana Chandra
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Peter A Kopp
- Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francis J Giles
- Northwestern Medicine Developmental Therapeutics Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
28
|
Pinto EM, Rodriguez-Galindo C, Pounds SB, Wang L, Clay MR, Neale G, Garfinkle EAR, Lam CG, Levy CF, Pappo AS, Zambetti GP, Ribeiro RC. Identification of Clinical and Biologic Correlates Associated With Outcome in Children With Adrenocortical Tumors Without Germline TP53 Mutations: A St Jude Adrenocortical Tumor Registry and Children's Oncology Group Study. J Clin Oncol 2017; 35:3956-3963. [PMID: 29058986 DOI: 10.1200/jco.2017.74.2460] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose The clinical features, pathogenesis, and outcomes in children with adrenocortical tumors (ACTs) without germline TP53 mutations have not been systematically studied. Herein, we describe these correlates and analyze their association with outcome. Patients and Methods Genomic DNA was analyzed for TP53, CTNNB1, CDKN1C, ATRX, and chromosome 11p15 abnormalities. β-catenin expression and Ki-67 labeling index (LI) were evaluated by immunostaining. Primary end points were progression-free (PFS) and overall survival. Results Median age of 42 girls and 18 boys was 3.3 years (range, 0.25 to 21.7 years). Complete resection (stages I and II) was achieved in 32 patients, and 28 patients had stage III or IV disease. Constitutional abnormalities of chromosome 11p15 occurred in nine of 40 patients, with six patients not showing phenotype of Beckwith-Wiedemann syndrome. Three-year PFS and overall survival for all patients were 71.4% and 80.5%, respectively. In single-predictor Cox regression analysis, age, disease stage, tumor weight, somatic TP53 mutations, and Ki-67 LI were associated with prognosis. Ki-67 LI and age remained significantly associated with PFS after adjusting for stage and tumor weight. Three-year PFS for 27 patients with Ki-67 LI ≥ 15% was 48.5% compared with 96.2% for 29 patients with Ki-67 LI < 15% (log-rank P = .002), and the rate of relapse increased by 24% with each 1-year increase in age at diagnosis (hazard ratio, 1.24; P = .0057). Conclusion Clinicopathologic features and outcomes of children with ACTs without germline TP53 mutations overlapped those reported for children with germline TP53 mutations. Our findings highlight the central role of genetic or epigenetic alterations on chromosome 11p15 in pediatric ACTs. Ki-67 LI is a strong prognostic indicator and should be investigated to improve the histologic classification of pediatric ACTs.
Collapse
Affiliation(s)
- Emilia Modolo Pinto
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Carlos Rodriguez-Galindo
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Stanley B Pounds
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Lei Wang
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Michael R Clay
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Geoffrey Neale
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Elizabeth A R Garfinkle
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Catherine G Lam
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Carolyn Fein Levy
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Alberto S Pappo
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Gerard P Zambetti
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| | - Raul C Ribeiro
- Emilia Modolo Pinto, Carlos Rodriguez-Galindo, Stanley B. Pounds, Lei Wang, Michael R. Clay, Geoffrey Neale, Catherine G. Lam, Alberto S. Pappo, Gerard P. Zambetti, and Raul C. Ribeiro, St Jude Children's Research Hospital; Elizabeth A.R. Garfinkle, University of Tennessee Health Science Center, Memphis, TN; and Carolyn Fein Levy, Steven and Alexandra Cohen Children's Medical Center, New York, NY
| |
Collapse
|
29
|
MacFarland SP, Mostoufi-Moab S, Zelley K, Mattei PA, States LJ, Bhatti TR, Duffy KA, Brodeur GM, Kalish JM. Management of adrenal masses in patients with Beckwith-Wiedemann syndrome. Pediatr Blood Cancer 2017; 64:10.1002/pbc.26432. [PMID: 28066990 PMCID: PMC5944603 DOI: 10.1002/pbc.26432] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/28/2016] [Accepted: 12/03/2016] [Indexed: 12/31/2022]
Abstract
Beckwith-Wiedemann syndrome (BWS) is a genetic overgrowth and cancer predisposition syndrome, associated with both benign and malignant adrenal findings. Literature review and an institutional case series elucidate the wide spectrum of adrenal findings in BWS patients. The altered expression of the 11p15 region is likely related to adrenal gland hyperplasia and growth dysregulation. Given the absence of guidelines for managing adrenal findings in BWS, we propose a systematic approach to adrenal findings in BWS patients, to allow for maximum detection of potentially malignant pathology without posing additional risk to patients.
Collapse
Affiliation(s)
- Suzanne P. MacFarland
- Division of Oncology, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Sogol Mostoufi-Moab
- Division of Oncology, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104,Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104
| | - Kristin Zelley
- Division of Oncology, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Peter A. Mattei
- Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104,Department of General, Thoracic, and Fetal Surgery, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Lisa J. States
- Department of Radiology, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Tricia R. Bhatti
- Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104,Department of Pathology and Laboratory Medicine, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Kelly A. Duffy
- Division of Human Genetics, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Garrett M. Brodeur
- Division of Oncology, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104,Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104
| | - Jennifer M. Kalish
- Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104,Division of Human Genetics, the Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
30
|
Babińska A, Pęksa R, Wiśniewski P, Świątkowska-Stodulska R, Sworczak K. Diagnostic and prognostic role of SF1, IGF2, Ki67, p53, adiponectin, and leptin receptors in human adrenal cortical tumors. J Surg Oncol 2017; 116:427-433. [PMID: 28672049 DOI: 10.1002/jso.24665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/23/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND The authors have examined the immunohistochemical expression of several proteins and their relationship with adrenal cortical carcinoma (ACC) diagnosis and progression. MATERIALS AND METHODS A total of 83 patients with benign and malignant adrenal cortex tumors operated on in a single center were included in the study. Expression of the following proteins was examined: steroidogenic factor 1 (SF1), insulin growth factor 2 (IGF2), Ki67, p53, as well as adiponectin (Adipo R1, Adipo R2), and leptin (Ob-R) receptors. RESULTS Multivariate analysis revealed that the expression of SF1, IGF2, and Adipo R1 and R2 receptors was associated with ACC diagnosis. An acknowledged proliferation marker Ki67 was related with the size of ACC and was an independent ACC diagnosis marker. The authors also assessed the relationship between immunohistochemical parameters and overall survival (OS) and disease progression. Only high IGF2 expression was associated with longer OS (P = 0.025). The most significant one for the prognosis of ACC patients was tumor resectability of the primary tumor. More favorable prognosis was found for young men (P = 0.033). CONCLUSIONS The presented data indicate that immunohistochemical assessment (of IGF2, SF1, Adipo R1, and R2 receptors' expression) may be useful in making the diagnosis of uncertain ACC cases.
Collapse
Affiliation(s)
- Anna Babińska
- Department of Endocrinology and Internal Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Rafał Pęksa
- Department of Pathology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Wiśniewski
- Department of Endocrinology and Internal Medicine, Medical University of Gdansk, Gdansk, Poland
| | | | - Krzysztof Sworczak
- Department of Endocrinology and Internal Medicine, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
31
|
Muscogiuri G, De Martino MC, Negri M, Pivonello C, Simeoli C, Orio F, Pivonello R, Colao A. Adrenal Mass: Insight Into Pathogenesis and a Common Link With Insulin Resistance. Endocrinology 2017; 158:1527-1532. [PMID: 28368448 DOI: 10.1210/en.2016-1804] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/24/2017] [Indexed: 12/29/2022]
Abstract
Adrenal mass (AM) is a common incidental finding detected during radiological investigations with an estimated incidence of 4%. Subjects with AM do not show any physical signs of adrenal hormonal excess, although they are often insulin resistant. Interestingly, apparently nonfunctioning AMs are often associated with a high prevalence of insulin resistance (IR) and metabolic syndrome. However, it is unclear whether AM develops from a primary IR and compensatory hyperinsulinemia or whether IR is only secondary to the slight cortisol hypersecretion by AM. Further, the degree of IR has been directly reported to correlate to the size of AM, thus allowing one to hypothesize that compensatory hyperinsulinemia to IR could be mitogenic on the adrenal cortex acting through the activation of insulin and insulinlike growth factor 1 receptors. Thus, the aim of the present article is to review the current evidence on the link between AM and compensatory hyperinsulinemia to IR.
Collapse
Affiliation(s)
| | | | | | - Claudia Pivonello
- Department of Clinical Medicine and Surgery, University of Naples, 80131 Naples, Italy
| | - Chiara Simeoli
- Department of Clinical Medicine and Surgery, University of Naples, 80131 Naples, Italy
| | - Francesco Orio
- Department of Sports Science and Wellness, "Parthenope" University Naples, 80131 Naples, Italy
| | - Rosario Pivonello
- Department of Clinical Medicine and Surgery, University of Naples, 80131 Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, University of Naples, 80131 Naples, Italy
| |
Collapse
|
32
|
Penny MK, Finco I, Hammer GD. Cell signaling pathways in the adrenal cortex: Links to stem/progenitor biology and neoplasia. Mol Cell Endocrinol 2017; 445:42-54. [PMID: 27940298 PMCID: PMC5508551 DOI: 10.1016/j.mce.2016.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/17/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
Abstract
The adrenal cortex is a dynamic tissue responsible for the synthesis of steroid hormones, including mineralocorticoids, glucocorticoids, and androgens in humans. Advances have been made in understanding the role of adrenocortical stem/progenitor cell populations in cortex homeostasis and self-renewal. Recently, large molecular profiling studies of adrenocortical carcinoma (ACC) have given insights into proteins and signaling pathways involved in normal tissue homeostasis that become dysregulated in cancer. These data provide an impetus to examine the cellular pathways implicated in adrenocortical disease and study connections, or lack thereof, between adrenal homeostasis and tumorigenesis, with a particular focus on stem and progenitor cell pathways. In this review, we discuss evidence for stem/progenitor cells in the adrenal cortex, proteins and signaling pathways that may regulate these cells, and the role these proteins play in pathologic and neoplastic conditions. In turn, we also examine common perturbations in adrenocortical tumors (ACT) and how these proteins and pathways may be involved in adrenal homeostasis.
Collapse
Affiliation(s)
- Morgan K Penny
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabella Finco
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gary D Hammer
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA; Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan Health System, 109 Zina Pitcher Place, 1528 BSRB, Ann Arbor, MI 48109, USA.
| |
Collapse
|
33
|
Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Minkevich AM, Rozanov AS, Peltek SE, Nikolin VP, Popova NA, Seledtsov IA, Molodtsov VV, Zavyalov EL, Taranov OS, Baiborodin SI, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Gene expression profiling of tumor-initiating stem cells from mouse Krebs-2 carcinoma using a novel marker of poorly differentiated cells. Oncotarget 2017; 8:9425-9441. [PMID: 28031533 PMCID: PMC5354742 DOI: 10.18632/oncotarget.14116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/15/2016] [Indexed: 12/18/2022] Open
Abstract
Using the ability of poorly differentiated cells to natively internalize fragments of extracellular double-stranded DNA as a marker, we isolated a tumorigenic subpopulation present in Krebs-2 ascites that demonstrated the features of tumor-inducing cancer stem cells. Having combined TAMRA-labeled DNA probe and the power of RNA-seq technology, we identified a set of 168 genes specifically expressed in TAMRA-positive cells (tumor-initiating stem cells), these genes remaining silent in TAMRA-negative cancer cells. TAMRA+ cells displayed gene expression signatures characteristic of both stem cells and cancer cells. The observed expression differences between TAMRA+ and TAMRA- cells were validated by Real Time PCR. The results obtained corroborated the biological data that TAMRA+ murine Krebs-2 tumor cells are tumor-initiating stem cells. The approach developed can be applied to profile any poorly differentiated cell types that are capable of immanent internalization of double-stranded DNA.
Collapse
Affiliation(s)
- Ekaterina A. Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Evgenia V. Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Anastasia S. Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yaroslav R. Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexandra M. Minkevich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Aleksey S. Rozanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey E. Peltek
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valeriy P. Nikolin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nelly A. Popova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | | | - Vladimir V. Molodtsov
- Novosibirsk State University, Novosibirsk 630090, Russia
- Softberry Inc., New York 10549, USA
| | - Evgeniy L Zavyalov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Oleg S. Taranov
- The State Research Center of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk 630559, Russia
| | - Sergey I. Baiborodin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Alexander A. Ostanin
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Elena R. Chernykh
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey S. Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
34
|
Creemers SG, van Koetsveld PM, van Kemenade FJ, Papathomas TG, Franssen GJH, Dogan F, Eekhoff EMW, van der Valk P, de Herder WW, Janssen JAMJL, Feelders RA, Hofland LJ. Methylation of IGF2 regulatory regions to diagnose adrenocortical carcinomas. Endocr Relat Cancer 2016; 23:727-37. [PMID: 27535174 DOI: 10.1530/erc-16-0266] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 01/20/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis. Discrimination of ACCs from adrenocortical adenomas (ACAs) is challenging on both imaging and histopathological grounds. High IGF2 expression is associated with malignancy, but shows large variability. In this study, we investigate whether specific methylation patterns of IGF2 regulatory regions could serve as a valuable biomarker in distinguishing ACCs from ACAs. Pyrosequencing was used to analyse methylation percentages in DMR0, DMR2, imprinting control region (ICR) (consisting of CTCF3 and CTCF6) and the H19 promoter. Expression of IGF2 and H19 mRNA was assessed by real-time quantitative PCR. Analyses were performed in 24 ACCs, 14 ACAs and 11 normal adrenals. Using receiver operating characteristic (ROC) analysis, we evaluated which regions showed the best predictive value for diagnosis of ACC and determined the diagnostic accuracy of these regions. In ACCs, the DMR0, CTCF3, CTCF6 and the H19 promoter were positively correlated with IGF2 mRNA expression (P<0.05). Methylation in the most discriminating regions distinguished ACCs from ACAs with a sensitivity of 96%, specificity of 100% and an area under the curve (AUC) of 0.997±0.005. Our findings were validated in an independent cohort of 9 ACCs and 13 ACAs, resulting in a sensitivity of 89% and a specificity of 92%. Thus, methylation patterns of IGF2 regulatory regions can discriminate ACCs from ACAs with high diagnostic accuracy. This proposed test may become the first objective diagnostic tool to assess malignancy in adrenal tumours and facilitate the choice of therapeutic strategies in this group of patients.
Collapse
Affiliation(s)
- S G Creemers
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - P M van Koetsveld
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - F J van Kemenade
- Department of PathologyErasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - T G Papathomas
- Department of PathologyErasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands Department of HistopathologyKing's College Hospital, Denmark Hill, London, UK
| | - G J H Franssen
- Department of SurgeryErasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - F Dogan
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - E M W Eekhoff
- Department of PathologyVU University Medical Center, Amsterdam, The Netherlands
| | - P van der Valk
- Department of PathologyVU University Medical Center, Amsterdam, The Netherlands
| | - W W de Herder
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J A M J L Janssen
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - R A Feelders
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - L J Hofland
- Department of Internal MedicineDivision of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
35
|
IGF2 knockdown in two colorectal cancer cell lines decreases survival, adhesion and modulates survival-associated genes. Tumour Biol 2016; 37:12485-12495. [PMID: 27337954 DOI: 10.1007/s13277-016-5115-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 06/09/2016] [Indexed: 12/22/2022] Open
Abstract
Increased expression of insulin-like growth factor 2 (IGF2) is found in tumors of colorectal cancer (CRC) patients exhibiting a gained region on chromosome 11q15 and is implicated in poor patient survival. This study analyzes in vitro phenotypic- and gene expression changes associated with IGF2 shRNA-mediated knockdown. Initially, doxycycline inducible IGF2 knockdown cell lines were generated in the CRC cell lines SW480 and LS174T. The cells were analyzed for changes in proliferation, cell cycle, apoptosis, adhesion, and invasion. Expression profiling analysis was performed, and, for a subset of the identified genes, expression was validated by qRT-PCR and Western blot. IGF2 knockdown inhibited cell proliferation in both cell lines induced G1 cell cycle blockade and decreased adhesion to several extracellular matrix proteins. Knockdown of IGF2 did not alter invasiveness in SW480 cells, while a slight increase in apoptosis was seen only in the LS174T cell line. Knockdown of IGF2 in SW480 deregulated 58 genes, several of which were associated with proliferation and cell-cell/cell-ECM contacts. A subset of these genes, including CDK2, YAP1, and BIRC5 (Survivin), are members of a common network. This study supports the concept of direct autocrine/paracrine tumor cell activation through IGF2 and a shows role of IGF2 in CRC proliferation, adhesion and, to a limited extent, apoptosis.
Collapse
|
36
|
Beuschlein F, Jakoby J, Mentz S, Zambetti G, Jung S, Reincke M, Süss R, Hantel C. IGF1-R inhibition and liposomal doxorubicin: Progress in preclinical evaluation for the treatment of adrenocortical carcinoma. Mol Cell Endocrinol 2016; 428:82-8. [PMID: 26994514 DOI: 10.1016/j.mce.2016.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 11/17/2022]
Abstract
Adrenocortical carcinoma (ACC) is a tumor with poor prognosis and limited therapeutic options. Therefore, in addition to multi-chemotherapeutic regimens IGF-1 receptor (IGF-1R) targeting approaches have been evaluated including immunoliposomal (IL) preparations utilizing an IGF-1R inhibiting antibody. In the current study, we extended our experiments by long-term treatment regimens in the classical adrenocortical NCIH295R xenograft model as well as by short-term experiments in two novel xenograft models, which all displayed different levels of IGF-1R and IGF-2 expression. Interestingly, these experiments reveal sub-group dependent differences in therapeutic outcome, reflecting clinical observations and indicate, thus, that implementation of this panel of tumor models might be helpful for clinical translation of novel therapeutic regimens in the future.
Collapse
Affiliation(s)
- Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Judith Jakoby
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Susanne Mentz
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerard Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sara Jung
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Reincke
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Adrenocortical carcinoma is a rare cancer, but one that carries a poor prognosis due to its aggressive nature and unresponsiveness to conventional chemotherapeutic strategies. Over the past 12 years, there has been renewed interest in developing new therapies for this cancer, including identifying key signaling nodes responsible for cell proliferation. RECENT FINDINGS Clinical trials of tyrosine kinase inhibitors as monotherapy have generally been disappointing, although the identification of exceptional responders may lead to the identification of targeted therapies that may produce responses in subsets of patients. Agents targeted to the Wnt signaling pathway, a known player in adrenal carcinogenesis, have been developed, although they have not yet been used specifically for adrenal cancer. There is current excitement about inhibitors of acetyl-coA cholesterol acetyl transferase 1, an enzyme required for intracellular cholesterol handling, although trials are still underway. Tools to target other proteins such as Steroidogenic Factor 1 and mechanistic target of rapamycin have been developed and are moving towards clinical application. SUMMARY Progress is being made in the fight against adrenocortical carcinoma with the identification of new therapeutic targets and new means by which to attack them. Continued improvement in the prognosis for patients with adrenal cancer is expected as this research continues.
Collapse
Affiliation(s)
- Bhavana Konda
- aDivision of OncologybDivision of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | | |
Collapse
|
38
|
Hantel C, Beuschlein F. Xenograft models for adrenocortical carcinoma. Mol Cell Endocrinol 2016; 421:28-33. [PMID: 26033247 DOI: 10.1016/j.mce.2015.05.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/30/2022]
Abstract
Adrenocortical carcinomas (ACCs) are rare, heterogeneous and very malignant endocrine tumors with a poor prognosis. An important prerequisite to optimize existing therapeutic regimens and to develop novel therapeutic strategies are preclinical disease models. In recent years molecular and genetic profiling of surgical tumor specimen led to the identification of novel interesting markers. However, precise involvement of these markers in tumorigenesis and their functional relevance in therapeutic outcome is still under investigation. Xenograft models are important tools for such functional studies as they bear the potential to mimic the complexity of solid tumors including tumor cells, stroma and blood vessels. Thus, for the successful and safe development of novel therapeutic strategies xenograft models remain to be indispensable experimental tools. Here we provide an overview on currently existing xenograft models for ACC, their tissue origins, establishment, implications as well as limitations.
Collapse
Affiliation(s)
- Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich, Germany.
| | - Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
39
|
Altieri B, Tirabassi G, Della Casa S, Ronchi CL, Balercia G, Orio F, Pontecorvi A, Colao A, Muscogiuri G. Adrenocortical tumors and insulin resistance: What is the first step? Int J Cancer 2015; 138:2785-94. [PMID: 26637955 DOI: 10.1002/ijc.29950] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/05/2015] [Accepted: 11/23/2015] [Indexed: 01/15/2023]
Abstract
The pathogenetic mechanisms underlying the onset of adrenocortical tumors (ACTs) are still largely unknown. Recently, more attention has been paid to the role of insulin and insulin-like growth factor (IGF) system on general tumor development and progression. Increased levels of insulin, IGF-1 and IGF-2 are associated with tumor cell growth and increased risk of cancer promotion and progression in patients with type 2 diabetes. Insulin resistance and compensatory hyperinsulinemia may play a role in adrenal tumor growth through the activation of insulin and IGF-1 receptors. Interestingly, apparently non-functioning ACTs are often associated with a high prevalence of insulin resistance and metabolic syndrome. However, it is unclear if ACT develops from a primary insulin resistance and compensatory hyperinsulinemia or if insulin resistance is only secondary to the slight cortisol hypersecretion by ACT. The aim of this review is to summarize the current evidence regarding the relationship between hyperinsulinemia and adrenocortical tumors.
Collapse
Affiliation(s)
- Barbara Altieri
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Giacomo Tirabassi
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Silvia Della Casa
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Cristina L Ronchi
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Giancarlo Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Orio
- Department of Sports Science and Wellness, Parthenope University, Naples, Italy.,Department of Endocrinology and Diabetology, Fertility Techniques Structure, University Hospital S. Giovanni Di Dio E Ruggi D'aragona, Salerno, Italy
| | - Alfredo Pontecorvi
- Institute of Medical Pathology, Division of Endocrinology and Metabolic Diseases, Catholic University, Rome, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Section of Endocrinology, Federico II University, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Clinical Medicine and Surgery, Section of Endocrinology, Federico II University, Naples, Italy
| |
Collapse
|
40
|
Nielsen HM, How-Kit A, Guerin C, Castinetti F, Vollan HKM, De Micco C, Daunay A, Taieb D, Van Loo P, Besse C, Kristensen VN, Hansen LL, Barlier A, Sebag F, Tost J. Copy number variations alter methylation and parallel IGF2 overexpression in adrenal tumors. Endocr Relat Cancer 2015; 22:953-67. [PMID: 26400872 PMCID: PMC4621769 DOI: 10.1530/erc-15-0086] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2015] [Indexed: 12/14/2022]
Abstract
Overexpression of insulin growth factor 2 (IGF2) is a hallmark of adrenocortical carcinomas and pheochromocytomas. Previous studies investigating the IGF2/H19 locus have mainly focused on a single molecular level such as genomic alterations or altered DNA methylation levels and the causal changes underlying IGF2 overexpression are still not fully established. In the current study, we analyzed 62 tumors of the adrenal gland from patients with Conn's adenoma (CA, n=12), pheochromocytomas (PCC, n=10), adrenocortical benign tumors (ACBT, n=20), and adrenocortical carcinomas (ACC, n=20). Gene expression, somatic copy number variation of chr11p15.5, and DNA methylation status of three differential methylated regions of the IGF2/H19 locus including the H19 imprinting control region were integratively analyzed. IGF2 overexpression was found in 85% of the ACCs and 100% of the PCCs compared to 23% observed in CAs and ACBTs. Copy number aberrations of chr11p15.5 were abundant in both PCCs and ACCs but while PCCs retained a diploid state, ACCs were frequently tetraploid (7/19). Loss of either a single allele or loss of two alleles of the same parental origin in tetraploid samples resulted in a uniparental disomy-like genotype. These copy number changes correlated with hypermethylation of the H19 ICR suggesting that the lost alleles were the unmethylated maternal alleles. Our data provide conclusive evidence that loss of the maternal allele correlates with IGF2 overexpression in adrenal tumors and that hypermethylation of the H19 ICR is a consequence thereof.
Collapse
Affiliation(s)
- Helene Myrtue Nielsen
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty
| | - Alexandre How-Kit
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Carole Guerin
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Frederic Castinetti
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Hans Kristian Moen Vollan
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty
| | - Catherine De Micco
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Antoine Daunay
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - David Taieb
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Peter Van Loo
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty
| | - Celine Besse
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Vessela N Kristensen
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty
| | - Lise Lotte Hansen
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Anne Barlier
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Frederic Sebag
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| | - Jörg Tost
- Laboratory for Functional GenomicsFondation Jean Dausset - Centre d'Etude du Polymorphisme Humain (CEPH), Paris, FranceInstitute of BiomedicineAarhus University, Aarhus, DenmarkEndocrine and Metabolic Surgery DepartmentAP-HM La Conception, Marseille, FranceDepartment of EndocrinologyAP-HM La Timone, Marseille, FranceDepartment of GeneticsInstitute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, NorwayDivision of SurgeryTransplantation and Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, NorwayThe K G Jebsen Center for Breast Cancer ResearchInstitute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, NorwayPathology DepartmentAP-HM La Timone, Marseille, FranceNuclear Endocrine Imaging and Treatment DepartmentAP-HM La Timone, Marseille, FranceCancer Research UKLondon Research Institute, London, UKDepartment of Human GeneticsUniversity of Leuven, Leuven, BelgiumGenotyping FacilitiesCentre National de Génotypage, CEA-Institut de Génomique, Evry, FranceDepartment of Clinical Molecular Biology (EpiGen)University of Oslo, Ahus, Lokerod, NorwayLaboratory of Molecular BiologyAP-HM La Conception and CRN2M, Aix-Marseille University, Marseille, FranceLaboratory for Epigenetics and EnvironmentCentre National de Génotypage, CEA-Institut de Génomique, Evry, France
| |
Collapse
|
41
|
Abstract
Advances in genomics accelerated greatly progress in the study of the genetics adrenocortical tumors. Bilateral nodular hyperplasias causing Cushing's syndrome are frequently caused by germline alterations leading to cAMP/PKA pathway activation (micronodular) and ARMC5 inactivation (macronodular). Somatic mutations of β-catenin and PRKACA are observed in non secreting or cortisol producing adenomas, respectively. Alterations of the β-catenin (CTNN1B, ZNFR3) or TP53 pathways are found in carcinomas. Mutations in cancers are more common in aggressive tumors and correlate with transcriptome or methylation profiles. Identification of these alterations helps to refine the molecular classification of these tumors and to develop molecular diagnostic tools.
Collapse
Affiliation(s)
- Stéphanie Espiard
- Cochin Institut, INSERM U1016, 24 rue du Faubourg Saint Jacques, Paris 75014, France; Cochin Institut, CNRS UMR8104, 24 rue du Faubourg Saint-Jacques, Paris 75014, France; Paris Descartes University, 12 rue de l'Ecole de Médecine, Paris 75006, France
| | - Jérôme Bertherat
- Cochin Institut, INSERM U1016, 24 rue du Faubourg Saint Jacques, Paris 75014, France; Cochin Institut, CNRS UMR8104, 24 rue du Faubourg Saint-Jacques, Paris 75014, France; Paris Descartes University, 12 rue de l'Ecole de Médecine, Paris 75006, France; Endocrinology Department, Center for Rare Adrenal Diseases, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, 27 Rue du Fg-St-Jacques, Paris F-75014, France.
| |
Collapse
|