151
|
Giordano TJ. Adrenocortical tumors: an integrated clinical, pathologic, and molecular approach at the University of Michigan. Arch Pathol Lab Med 2010; 134:1440-3. [PMID: 20923297 DOI: 10.5858/2010-0320-ra.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT The University of Michigan Health System has a rich tradition in the study and treatment of endocrine neoplasia. Recently, an integrated clinical and research program focused on primary cancer of the adrenal gland has been developed. OBJECTIVE To discuss the foundation of the University of Michigan Adrenal Cancer Program that consists of 3 components: (1) advancement of excellent clinical care, (2) dissemination of resources and knowledge, and (3) scientific discovery. DATA SOURCES Recent programmatic activity includes genome-wide transcriptomic evaluation of human adrenocortical tumors for diagnostic and prognostic evaluation; interrogation of the Wnt signaling pathway in adrenocortical carcinoma, using mouse models and transcriptome profiling; and clinical trials with targeted therapy focused on inhibition of insulin-like growth factor signaling pathway. CONCLUSIONS This review article presents an overview of this program and its recent activity.
Collapse
Affiliation(s)
- Thomas J Giordano
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109-0669, USA.
| |
Collapse
|
152
|
Patterson EE, Holloway AK, Weng J, Fojo T, Kebebew E. MicroRNA profiling of adrenocortical tumors reveals miR-483 as a marker of malignancy. Cancer 2010; 117:1630-9. [PMID: 21472710 DOI: 10.1002/cncr.25724] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/09/2010] [Accepted: 09/13/2010] [Indexed: 11/07/2022]
Abstract
BACKGROUND The authors are interested in identifying molecular markers that can aid in the diagnosis of adrenocortical carcinoma (ACC). The aim of this study was to identify microRNAs (miRNAs or miRs) that are differentially expressed in malignant adrenocortical tumors as compared with benign tumors and assess their potential as diagnostic predictors. METHODS Differentially expressed miRNAs were identified using microarray profiling of adrenocortical tumors and validated by quantitative real-time RT-PCR. RESULTS Microarray profiling in benign and primary malignant adrenocortical tumors revealed several significant differences between these histological groups. By using directed quantitative RT-PCR analysis on a subset of these differentially expressed miRNAs, the authors determined that miRs -100, -125b, and -195 were significantly down-regulated, whereas miR-483-5p was significantly up-regulated in malignant as compared with benign tumors. Furthermore, the current study shows that miR-483-5p expression can accurately categorize tumors as benign or malignant. CONCLUSIONS The authors identified 4 miRNAs that are dysregulated in adrenocortical carcinoma. The high expression of one of these, miR-483-5p, appears to be a defining characteristic of adrenocortical malignancies, and can thus be used to accurately distinguish between benign and malignant adrenocortical tumors.
Collapse
Affiliation(s)
- Erin E Patterson
- Endocrine Oncology Section, Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
153
|
Gaujoux S, Pinson S, Gimenez-Roqueplo AP, Amar L, Ragazzon B, Launay P, Meatchi T, Libé R, Bertagna X, Audebourg A, Zucman-Rossi J, Tissier F, Bertherat J. Inactivation of the APC gene is constant in adrenocortical tumors from patients with familial adenomatous polyposis but not frequent in sporadic adrenocortical cancers. Clin Cancer Res 2010; 16:5133-41. [PMID: 20978149 DOI: 10.1158/1078-0432.ccr-10-1497] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE In adrenocortical tumors (ACT), Wnt/β-catenin pathway activation can be explained by β-catenin somatic mutations only in a subset of tumors. ACT is observed in patients with familial adenomatous polyposis (FAP) with germline APC mutations, as well as in patients with Beckwith-Wiedemann syndrome with Wilms' tumors reported to have WTX somatic mutations. Both APC and WTX are involved in Wnt/β-catenin pathway regulation and may play a role in ACT tumorigenesis. The aim of this study was to report if APC and WTX may be associated with FAP-associated and sporadic ACT. EXPERIMENTAL DESIGN ACTs from patients with FAP and sporadic adrenocortical carcinomas (ACC) with abnormal β-catenin localization on immunohistochemistry but no somatic β-catenin mutations were studied. APC was analyzed by denaturing high-performance liquid chromatography followed by direct sequencing and by multiplex ligation-dependent probe amplification when allelic loss was suspected. WTX was studied by direct sequencing. RESULTS Four ACTs were observed in three patients with FAP and were ACC, adrenocortical adenoma, and bilateral macronodular adrenocortical hyperplasia, all with abnormal β-catenin localization. Biallelic inactivation of APC was strongly suggested by the simultaneous existence of somatic and germline alterations in all ACTs. In the 20 sporadic ACCs, a silent heterozygous somatic mutation as well as a rare heterozygous polymorphism in APC was found. No WTX mutations were observed. CONCLUSIONS ACT should be considered a FAP tumor. Biallelic APC inactivation mediates activation of the Wnt/β-catenin pathway in the ACTs of patients with FAP. In contrast, APC and WTX genetic alterations do not play a significant role in sporadic ACC.
Collapse
Affiliation(s)
- Sébastien Gaujoux
- Institut Cochin, Université Paris Descartes-Faculté de médecine, CNRS (UMR 8104), Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Ragazzon B, Libé R, Gaujoux S, Assié G, Fratticci A, Launay P, Clauser E, Bertagna X, Tissier F, de Reyniès A, Bertherat J. Transcriptome analysis reveals that p53 and {beta}-catenin alterations occur in a group of aggressive adrenocortical cancers. Cancer Res 2010; 70:8276-81. [PMID: 20959480 DOI: 10.1158/0008-5472.can-10-2014] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare disease with an overall poor but heterogeneous prognosis. This heterogeneity could reflect different mechanisms of tumor development. Gene expression profiling by transcriptome analysis led to ACC being divided into two groups of tumors with very different outcomes. Somatic inactivating mutations of the tumor suppressor gene TP53 and activating mutations of the proto-oncogene β-catenin (CTNNB1) are the most frequent mutations identified in ACC. This study investigates the correlation between p53 and β-catenin alterations and the molecular classification of ACC by transcriptome analysis of 51 adult sporadic ACCs. All TP53 and CTNNB1 mutations seemed to be mutually exclusive and were observed only in the poor-outcome ACC group. Most of the abnormal p53 and β-catenin immunostaining was also found in this group. Fifty-two percent of the poor-outcome ACC group had TP53 or CTNNB1 mutations and 60% had abnormal p53 or β-catenin immunostaining. Unsupervised clustering transcriptome analysis of this poor-outcome group revealed three different subgroups, two of them being associated with p53 or β-catenin alterations, respectively. Analysis of p53 and β-catenin target gene expressions in each cluster confirmed a profound and anticipated effect on tumor biology, with distinct profiles logically associated with the respective pathway alterations. The third group had no p53 or β-catenin alteration, suggesting other unidentified molecular defects. This study shows the important respective roles of p53 and β-catenin in ACC development, delineating subgroups of ACC with different tumorigenesis and outcomes.
Collapse
Affiliation(s)
- Bruno Ragazzon
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, Inserm, U1016, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
McNicol AM. Update on tumours of the adrenal cortex, phaeochromocytoma and extra-adrenal paraganglioma. Histopathology 2010; 58:155-68. [DOI: 10.1111/j.1365-2559.2010.03613.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
156
|
Proposal for modification of the ENSAT staging system for adrenocortical carcinoma using tumor grade. Langenbecks Arch Surg 2010; 395:955-61. [PMID: 20694732 DOI: 10.1007/s00423-010-0698-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Accepted: 07/12/2010] [Indexed: 01/18/2023]
Abstract
PURPOSE Various staging systems for adrenocortical carcinoma (ACC) have been proposed. We hypothesized that incorporating tumor grade into the current European Network for the Study of Adrenal Tumors (ENSAT) staging system would improve the ability to more accurately predict time to recurrence and death. METHODS A retrospective review of patients included in the University of Michigan ACC database from 2005 to 2009 was done; and stage, tumor grade, time to recurrence, and death were recorded and analyzed using the Cox regression and Kaplan-Meier survival curves. RESULTS Ninety one patients had complete information for inclusion. The median follow-up was 24 months while the median time to recurrence was 4.1 months. There were 28 deaths; overall, tumor grade showed a significant difference in time to tumor recurrence (p = 0.011) and time to death (p = 0.004). Time to death among stage 2 patients separated into those with high- and low-grade tumors reached statistical significance (p = 0.05), and notable but not statistically significant differences were identified in all stages. Based on tumor grade and survival curves, modifications to the current ENSAT staging system were made. CONCLUSION Tumor grade plays a significant role in the outcome of patients with ACC. High-grade tumors are associated with shorter disease-free intervals and shorter overall survival. The proposed modification of the ENSAT staging system allows for incorporation of tumor grade when predicting overall survival.
Collapse
|
157
|
Ye L, Santarpia L, Gagel RF. The evolving field of tyrosine kinase inhibitors in the treatment of endocrine tumors. Endocr Rev 2010; 31:578-99. [PMID: 20605972 DOI: 10.1210/er.2009-0031] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Activation of tyrosine kinase receptors (TKRs) and their related pathways has been associated with development of endocrine tumors. Compounds that target and inactivate the kinase function of these receptors, tyrosine kinase inhibitors (TKIs), are now being applied to the treatment of endocrine tumors. Recent clinical trials of TKIs in patients with advanced thyroid cancer, islet cell carcinoma, and carcinoid have shown promising preliminary results. Significant reductions in tumor size have been described in medullary and papillary thyroid carcinoma, although no complete responses have been reported. Case reports have described significant tumor volume reductions of malignant pheochromocytomas and paragangliomas. In addition, these compounds showed an initial tumoricidal or apoptotic response followed by long-term static effects on tumor growth. Despite the promising preliminary results, this class of therapeutic agents has a broad spectrum of adverse effects, mediated by inhibition of kinase activities in normal tissues. These adverse effects will have to be balanced with their benefit in clinical use. New strategies will have to be applied in clinical research to achieve optimal benefits. In this review, we will address the genetic alterations of TKRs, the rationale for utilizing TKIs for endocrine tumors, and current information on tumor and patient responses to specific TKIs. We will also discuss the adverse effects related to TKI treatment and the mechanisms involved. Finally, we will summarize the challenges associated with use of this class of compounds and potential solutions.
Collapse
Affiliation(s)
- Lei Ye
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M. D. Anderson Cancer Center, Houston, 77030, USA
| | | | | |
Collapse
|
158
|
Szabó D, Zsippai A, Bendes M, Tömböl Z, Szabó PM, Rácz K, Igaz P. Pathogenesis of adrenocortical cancer. Orv Hetil 2010; 151:1163-70. [DOI: 10.1556/oh.2010.28931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A mellékvesekéreg-carcinoma ritka, rossz prognózisú daganat. Döntően sporadikus előfordulású, de ismertek nagyon ritka öröklődő formái is, amelyek a patogenezis megértésében nagy segítséget nyújtanak. A mellékvesekéreg-daganatokra hajlamosító öröklődő szindrómák közé tartozik a Li–Fraumeni-szindróma, a Beckwith–Wiedemann-szindróma, a familiáris adenomatosus polyposis, illetve a döntően benignus daganatokkal társuló multiplex endokrin neoplasia 1-es típusa (MEN1), Carney-komplex és McCune–Albright-szindróma. A mellékvesekéreg-daganatok patogenezisében szereplő főbb mechanizmusok közé tartozik az inzulinszerű növekedési faktor-2 fokozott expressziója, a Wnt/β-katenin és a cAMP-proteinkináz-A jelátviteli utak aktivációja, valamint a p53 és MEN1 gének mutációi. A mellékvesekéreg-carcinoma kezelésében a gyógyszeres lehetőségek meglehetősen korlátozottak. Az utóbbi évek molekuláris-bioinformatikai kutatásai számos eddig ismeretlen patogenetikai út szerepét vetették fel, amelyek új gyógyszeres támadáspontok lehetőségét is jelenthetik. E tanulmányban a szerzők az öröklődő daganatszindrómák patogenezisét, a sporadikus daganatokban észlelt eltéréseket és a legújabb molekuláris-bioinformatikai eredményeket ismertetik.
Collapse
Affiliation(s)
- Diána Szabó
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Adrienn Zsippai
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Melinda Bendes
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Zsófia Tömböl
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Péter M. Szabó
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Károly Rácz
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Péter Igaz
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| |
Collapse
|
159
|
Soon PS, Tacon LJ, Clifton-Bligh RJ, Robinson BG, Sidhu SB, Campbell PR, Gill AJ, Bambach CP, Sywak MS, Yeh MW, Wong SG. MicroRNA Microarray Analysis of Human Adrenocortical Tumors Identifies miR-195 and miR-483-5p as Predictors of Poor Prognosis in Adrenocortical Cancer – Response. Clin Cancer Res 2010. [DOI: 10.1158/1078-0432.ccr-10-0594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Patsy S.H. Soon
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Lyndal J. Tacon
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Roderick J. Clifton-Bligh
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Bruce G. Robinson
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Stan B. Sidhu
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Peter R. Campbell
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Anthony J. Gill
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Christopher P. Bambach
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Mark S. Sywak
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Michael W. Yeh
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| | - Steven G. Wong
- 1Cancer Genetics, Kolling Institute of Medical Research and University of Sydney, Sydney, Australia 2Department of Surgery, Bankstown Hospital and South Western Sydney Clinical School, University of New South Wales, Sydney, Australia 3Department of Endocrinology, Royal North Shore Hospital, Sydney, Australia 4Department of Anatomical Pathology, Royal North Shore Hospital, and University of Sydney, Sydney, Australia 5University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia 6Department of Surgery, Liverpool Hospital, Sydney, Australia 7Endocrine Surgical Unit, University of California Los Angeles, Los Angeles, California 8Department of Medicine, Division of Hematology and Oncology, University of California at Los Angeles School of Medicine, Los Angeles, California
| |
Collapse
|
160
|
Assié G, Guillaud-Bataille M, Ragazzon B, Bertagna X, Bertherat J, Clauser E. The pathophysiology, diagnosis and prognosis of adrenocortical tumors revisited by transcriptome analyses. Trends Endocrinol Metab 2010; 21:325-34. [PMID: 20097573 DOI: 10.1016/j.tem.2009.12.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 12/15/2009] [Accepted: 12/18/2009] [Indexed: 11/24/2022]
Abstract
Analyzing gene expression (transcriptome) in tissue is now reliable using industrial pangenomic microarrays. Accumulating data on adrenal cortex and adrenocortical tumor transcriptomes have already identified striking transcriptome differences not only between adenoma and carcinoma but also between two sets of carcinoma, which have very different prognoses. These findings result in the development of diagnostic and prognostic molecular predictors, which improve the outcome determination compared with standard clinical and pathological tools. These transcriptome data observing adrenocortical tumor phenotype in great but complex detail, combined with genomic and proteomic information, will function for future research investigating the pathophysiology of their tumorigenesis and hormonal secretion.
Collapse
Affiliation(s)
- Guillaume Assié
- Department of Endocrinology, Metabolism and Cancer, Institut Cochin, INSERM U567, University Paris Descartes, CNRS UMR8104, Paris, France
| | | | | | | | | | | |
Collapse
|
161
|
Ishikura K, Takamura T, Takeshita Y, Nakagawa A, Imaizumi N, Misu H, Taji K, Kasahara K, Oshinoya Y, Suzuki S, Ooi A, Kaneko S. Cushing's syndrome and big IGF-II associated hypoglycaemia in a patient with adrenocortical carcinoma. BMJ Case Rep 2010; 2010:bcr07.2009.2100. [PMID: 22461853 DOI: 10.1136/bcr.07.2009.2100] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A 41-year-old woman had a general health examination and was diagnosed with a non-functioning adrenocortical carcinoma (ACC). Despite surgery and chemotherapy with mitotane, the ACC progressed with metastases to the lymph nodes, liver and lung. Initially, she developed adrenal insufficiency and was treated with hydrocortisone. As the ACC progressed, it produced superabundant cortisol, resulting in clinically overt Cushing's syndrome. As the liver metastases grew, the patient developed hypoglycaemia with suppression of endogenous insulin secretion. She had to be given large quantities of glucose intravenously to remain normoglycaemic. The serum insulin-like growth factor (IGF)-II/IGF-I ratio had increased to 84. We identified big IGF-II, a primary hormonal mediator of non-islet cell tumour hypoglycaemia (NICTH), in the serum and tumour using western blotting. This is the first case of ACC that showed both Cushing's syndrome and NICTH associated with big IGF-II.
Collapse
Affiliation(s)
- Kazuhide Ishikura
- Kanazawa University Graduate School of Medical Science, Department of Disease Control and Homeostasis, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Meta-analysis of adrenocortical tumour genomics data: novel pathogenic pathways revealed. Oncogene 2010; 29:3163-72. [DOI: 10.1038/onc.2010.80] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
163
|
Looyenga BD, Wiater E, Vale W, Hammer GD. Inhibin-A antagonizes TGFbeta2 signaling by down-regulating cell surface expression of the TGFbeta coreceptor betaglycan. Mol Endocrinol 2010; 24:608-20. [PMID: 20160125 DOI: 10.1210/me.2008-0374] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inhibin is an atypical member of the TGFbeta family of signaling ligands and is classically understood to function via competitive antagonism of activin ligand binding. Inhibin-null (Inha-/-) mice develop both gonadal and adrenocortical tumors, the latter of which depend upon gonadectomy for initiation. We have previously shown that gonadectomy initiates adrenal tumorigenesis in Inha-/- mice by elevating production of LH, which drives aberrant proliferation and differentiation of subcapsular adrenocortical progenitor cells. In this study, we demonstrate that LH signaling specifically up-regulates expression of TGFbeta2 in the subcapsular region of the adrenal cortex, which coincides with regions of aberrant Smad3 activation in Inha-/- adrenal glands. Consistent with a functional interaction between inhibin and TGFbeta2, we further demonstrate that recombinant inhibin-A antagonizes signaling by TGFbeta2 in cultured adrenocortical cells. The mechanism of this antagonism depends upon the mutual affinity of inhibin-A and TGFbeta2 for the signaling coreceptor betaglycan. Although inhibin-A cannot physically displace TGFbeta2 from its binding sites on betaglycan, binding of inhibin-A to the cell surface causes endocytic internalization of betaglycan, thereby reducing the number of available binding sites for TGFbeta2 on the cell surface. The mechanism by which inhibin-A induces betaglycan internalization is clathrin independent, making it distinct from the mechanism by which TGFbeta ligands themselves induce betaglycan internalization. These data indicate that inhibin can specifically antagonize TGFbeta2 signaling in cellular contexts where surface expression of betaglycan is limiting and provide a novel mechanism for activin-independent phenotypes in Inha-/- mice.
Collapse
|
164
|
Berthon A, Sahut-Barnola I, Lambert-Langlais S, de Joussineau C, Damon-Soubeyrand C, Louiset E, Taketo MM, Tissier F, Bertherat J, Lefrançois-Martinez AM, Martinez A, Val P. Constitutive beta-catenin activation induces adrenal hyperplasia and promotes adrenal cancer development. Hum Mol Genet 2010; 19:1561-76. [PMID: 20106872 DOI: 10.1093/hmg/ddq029] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Adrenocortical carcinoma is a rare but aggressive cancer with unknown aetiology. Constitutive activation of beta-catenin is the most frequent alteration in benign and malignant adrenocortical tumours in patients. Here, we show that constitutive activation of beta-catenin in the adrenal cortex of transgenic mice resulted in progressive steroidogenic and undifferentiated spindle-shaped cells hyperplasia as well as dysplasia of the cortex and medulla. Over a 17 months time course, transgenic adrenals developed malignant characteristics such as uncontrolled neovascularization and loco-regional metastatic invasion. These oncogenic events were accompanied by ectopic differentiation of glomerulosa at the expense of fasciculata cells, which caused primary hyperaldosteronism. Altogether these observations demonstrate that constitutively active beta-catenin is an adrenal oncogene which triggers benign aldosterone-secreting tumour development and promotes malignancy.
Collapse
Affiliation(s)
- Annabel Berthon
- Clermont Université, Université Blaise Pascal, GReD, BP 10448, F-63000 Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
|
166
|
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy causing up to 0.2% of all cancer deaths This article reviews the incidence, presentation, and pathology of ACC. Particular attention is paid to the molecular oncogenesis of this disease, and the surgical and therapeutic options available for its cure.
Collapse
Affiliation(s)
- Melissa Wandoloski
- Translational Genomics Research Institute, Clinical Translational Research Division, Phoenix, AZ 85004, USA
| | | | | |
Collapse
|
167
|
Crand A, Borson-Chazot F, Brue T. [Recent data in adrenocortical tumorigenesis]. ANNALES D'ENDOCRINOLOGIE 2009; 70 Suppl 1:S20-5. [PMID: 19878765 DOI: 10.1016/s0003-4266(09)72472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Adrenocortical carcinomas are rare tumors characterized by an aggressive behaviour with a 5-year survival rate below 30%. Until now, surgery is the only curative treatment for tumors confined to the adrenal gland and there is a lack of an effective medical treatment for invasive or metastatic tumors due to the poor knowledge of the mechanisms underlying adrenocortical malignancy. Moreover, histopathology is sometimes insufficient to establish an accurate diagnosis between a benign and a malignant adrenal tumor and a poor indicator of prognosis. In the last decade, the study of rare genetic syndromes associated with adrenocortical carcinomas and the identification of genetic alterations in adrenal tumors has improved our understanding of the pathogenesis of adrenal tumors. The development of molecular predictors of malignancy and of survival could help for histological diagnosis and determination of prognosis. These significant advances are essential to improve adrenocortical carcinoma management. This review summarizes recent advances in the understanding and management of adrenocortical tumors.
Collapse
Affiliation(s)
- A Crand
- Fédération d'endocrinologie du pôle Est, Hospices Civils de Lyon, 59, Boulevard Pinel, 69677 Bron.
| | | | | |
Collapse
|
168
|
Tacon LJ, Soon PS, Gill AJ, Chou AS, Clarkson A, Botling J, Stalberg PLH, Skogseid BM, Robinson BG, Sidhu SB, Clifton-Bligh RJ. The glucocorticoid receptor is overexpressed in malignant adrenocortical tumors. J Clin Endocrinol Metab 2009; 94:4591-9. [PMID: 19820023 DOI: 10.1210/jc.2009-0546] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
CONTEXT Adrenocortical carcinoma (ACC) is a rare tumor with a poor prognosis. The Weiss score is the most widely accepted method for distinguishing an ACC from an adrenocortical adenoma (ACA); however, in borderline cases, accurate diagnosis remains problematic. We recently discovered that the glucocorticoid receptor (GR) gene NR3C1 is significantly up-regulated in ACCs compared with ACAs in global gene expression studies. OBJECTIVE Our objective was to study GR expression in adrenocortical tumors (ACTs) and to assess its utility as an adjunct to the Weiss score. DESIGN Microarray analysis, real-time quantitative RT-PCR (qPCR), immunohistochemistry, Western blot, and direct sequencing were performed. RESULTS Analysis of 28 ACTs by microarray and 49 ACTs by qPCR found NR3C1 expression to be up-regulated in ACCs compared with ACAs (P < 0.001). Western blotting and RT-PCR confirmed the presence of the GRalpha isoform in ACCs, and no mutations were detected on direct sequencing. Immunohistochemistry for GR in an overlapping cohort of ACTs demonstrated strongly positive nuclear staining in 31 of 33 ACCs (94%), with negative staining in 40 of 41 ACAs (98%) (P < 0.001). This finding was validated in an external cohort of ACTs, such that 14 of 18 ACCs (78%) demonstrated positive nuclear staining whereas 32 of 33 ACAs (94%) were negative (P < 0.001). CONCLUSIONS The immunohistochemical finding of nuclear GR staining identified ACCs with high diagnostic accuracy. We propose that GR immunohistochemistry may complement the Weiss score in the diagnosis of ACC in cases that display borderline histology. The possibility that GR is transcriptionally active in these tumors, and may therefore be a therapeutic target, requires further study.
Collapse
Affiliation(s)
- Lyndal J Tacon
- Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2006, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, Paccagnella ML, de Bono JS, Gualberto A, Hammer GD. Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol 2009; 65:765-73. [PMID: 19649631 DOI: 10.1007/s00280-009-1083-9] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Accepted: 07/12/2009] [Indexed: 01/19/2023]
Abstract
PURPOSE Insulin-like growth factor 1 receptor signaling through upregulation of the stimulatory ligand IGF-II has been implicated in the pathogenesis of adrenocortical carcinoma. As there is a paucity of effective therapies, this dose expansion cohort of a phase 1 study was undertaken to determine the safety, tolerability, pharmacokinetics, and effects on endocrine markers of figitumumab in patients with adrenocortical carcinoma. METHODS Figitumumab was administered on day 1 of each 21-day cycle at the maximal feasible dose (20 mg/kg) to a cohort of patients with metastatic, refractory adrenocortical carcinoma. Serum glucose, insulin, and growth hormone were measured pre-study, at cycle 4 and study end. Pharmacokinetic evaluation was performed during cycles 1 and 4. RESULTS Fourteen patients with adrenocortical carcinoma received 50 cycles of figitumumab at the 20 mg/kg. Treatment-related toxicities were generally mild and included hyperglycemia, nausea, fatigue, and anorexia. Single episodes of grade 4 hyperuricemia, proteinuria, and elevated gamma-glutamyltransferase were observed. Pharmacokinetics of figitumumab was comparable to patients with solid tumors other than adrenocortical carcinoma. Treatment with figitumumab increased serum insulin and growth hormone levels. Eight of 14 patients (57%) had stable disease. CONCLUSIONS The side effect profile and pharmacokinetics of figitumumab were similar in patients with adrenocortical carcinoma in comparison to patients with other solid tumors. While hyperglycemia was the most common adverse event, no clear patterns predicting severity were observed. The majority of patients receiving protocol therapy with single agent figitumumab experienced stability of disease, warranting further evaluation.
Collapse
Affiliation(s)
- Paul Haluska
- Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Moulin C, Metzger-Filho O, Awada A. Changing the clinical picture of challenging tumors: tales becoming reality? Future Oncol 2009; 5:785-802. [DOI: 10.2217/fon.09.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The treatment of neoplastic diseases has become increasingly dependent on tumor biology and is focused on targeted therapy. Understanding complex networks of intracellular signaling pathways, blockades of specific targets and a myriad of other approaches has brought new fuel to the battle against many types of cancer. Unfortunately, the degree of benefit achieved in this new era of cancer treatment has not been distributed homogeneously among the different disease types. Neoplasms with lower incidence rates, but that are also highly challenging, are not consistently given due attention by research leaders. This article aims to evaluate new insights and potential gains obtained with new therapies in a particular group of tumors: those rarely debated in clinical practice, but which still pose a considerable challenge to clinical oncology.
Collapse
Affiliation(s)
- Camilo Moulin
- Jules Bordet Institute, 121 Bd. de Waterloo, 1000 Brussels, Belgium
| | | | - Ahmad Awada
- Jules Bordet Institute, 121 Bd. de Waterloo, 1000 Brussels, Belgium
| |
Collapse
|
171
|
Giordano TJ, Kuick R, Else T, Gauger PG, Vinco M, Bauersfeld J, Sanders D, Thomas DG, Doherty G, Hammer G. Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clin Cancer Res 2009; 15:668-76. [PMID: 19147773 DOI: 10.1158/1078-0432.ccr-08-1067] [Citation(s) in RCA: 270] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Our understanding of adrenocortical carcinoma (ACC) has improved considerably, yet many unanswered questions remain. For instance, can molecular subtypes of ACC be identified? If so, what is their underlying pathogenetic basis and do they possess clinical significance? EXPERIMENTAL DESIGN We did a whole genome gene expression study of a large cohort of adrenocortical tissues annotated with clinicopathologic data. Using Affymetrix Human Genome U133 Plus 2.0 oligonucleotide arrays, transcriptional profiles were generated for 10 normal adrenal cortices (NC), 22 adrenocortical adenomas (ACA), and 33 ACCs. RESULTS The overall classification of adrenocortical tumors was recapitulated using principal component analysis of the entire data set. The NC and ACA cohorts showed little intragroup variation, whereas the ACC cohort revealed much greater variation in gene expression. A robust list of 2,875 differentially expressed genes in ACC compared with both NC and ACA was generated and used in functional enrichment analysis to find pathways and attributes of biological significance. Cluster analysis of the ACCs revealed two subtypes that reflected tumor proliferation, as measured by mitotic counts and cell cycle genes. Kaplan-Meier analysis of these ACC clusters showed a significant difference in survival (P < 0.020). Multivariate Cox modeling using stage, mitotic rate, and gene expression data as measured by the first principal component for ACC samples showed that gene expression data contains significant independent prognostic information (P < 0.017). CONCLUSIONS This study lays the foundation for the molecular classification and prognostication of adrenocortical tumors and also provides a rich source of potential diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Thomas J Giordano
- Department of Pathology, 1150 West Medical Center Drive, MSRB-2, C570D, Ann Arbor, MI 48109-0669, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Johnsen IK, Kappler R, Auernhammer CJ, Beuschlein F. Bone morphogenetic proteins 2 and 5 are down-regulated in adrenocortical carcinoma and modulate adrenal cell proliferation and steroidogenesis. Cancer Res 2009; 69:5784-92. [PMID: 19584291 DOI: 10.1158/0008-5472.can-08-4428] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bone morphogenetic proteins (BMP) have been shown to affect tumorigenesis in a variety of tumors. Quantitative PCR analysis revealed down-regulation of BMP2 and BMP5 in tissue samples from adrenocortical carcinoma and adrenocortical tumor cell lines compared with normal adrenal glands. Integrity of BMP-dependent pathways in these cell lines could be shown by activation of the Smad1/5/8 pathway with subsequent increase of ID protein expression upon incubation with BMP2 or BMP5. On a functional level, BMP treatment resulted in inhibition of cell proliferation and viability in a dose- and time-dependent manner. This growth inhibitory effect was associated with BMP-dependent reduction of AKT phosphorylation under baseline conditions and under insulin-like growth factor costimulation. Furthermore, steroidogenic function, including melanocortin-2 receptor and steroidogenic enzyme expressions, was profoundly reduced. In vitro demethylation treatment and overexpression of GATA6 resulted in reactivation of BMP-dependent pathways with concomitant modulation of steroidogenesis. Taken together, we show that loss of expression of members of the BMP family of ligands is a common finding in adrenocortical tumors and we provide evidence that BMP-dependent pathways are likely to be involved in the modulation of the malignant and functional phenotype of adrenocortical cancer cells.
Collapse
Affiliation(s)
- Inga K Johnsen
- Departments of Medicine, University Hospital Innenstadt, Ludwig Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
173
|
Weismann D, Briese J, Niemann J, Grüneberger M, Adam P, Hahner S, Johanssen S, Liu W, Ezzat S, Saeger W, Bamberger AM, Fassnacht M, Schulte HM, Asa SL, Allolio B, Bamberger CM. Osteopontin stimulates invasion of NCI-h295 cells but is not associated with survival in adrenocortical carcinoma. J Pathol 2009; 218:232-40. [PMID: 19326399 DOI: 10.1002/path.2528] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Gene array studies indicated that osteopontin (OPN) mRNA is highly expressed in adrenocortical carcinomas (ACCs). OPN enhances invasiveness, proliferation, and metastasis formation, and is associated with poor survival in some malignant diseases. Integrin alphavbeta3 has been shown to mediate OPN effects on invasion. In this study, we demonstrated OPN and integrin alphavbeta3 expression in normal adrenal glands and benign adenomas, with staining seen exclusively in adrenocortical cells as well as even stronger staining in ACC. Western blot analysis confirmed overexpression of OPN in ACC (p < 0.01). With Matrigel invasion assays, we have shown that OPN greatly stimulates the invasiveness of NCI-h295 cells (>six-fold increase, p < 0.001). Transfection with integrin alphavbeta3 further increased invasiveness after OPN stimulation (p < 0.001). This increase was reversed by the addition of an anti-integrin beta3 antibody, indicating a functional relationship of OPN and integrin alphavbeta3 in ACC. With tissue arrays, we confirmed high OPN expression in 147 ACC samples. However, no association with survival was seen in Kaplan-Meier analysis including 111 patients with primary tumours graded for OPN staining and follow-up data available. In conclusion, our in vitro data indicate that OPN and integrin alphavbeta3 may act as a functional complex facilitating the invasiveness of adrenocortical tumours. This relationship remains of relevance to our understanding of carcinogenesis, but further studies are needed to address the physiological and pathological function of OPN in adrenal tissue.
Collapse
Affiliation(s)
- Dirk Weismann
- University Hospital of Würzburg, Department of Internal Medicine I, Endocrine and Diabetes Unit, Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Tissier F. Anatomie pathologique des tumeurs corticosurrénaliennes de l’adulte : état des lieux et données récentes. ANNALES D'ENDOCRINOLOGIE 2009; 70:179-85. [DOI: 10.1016/j.ando.2009.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
175
|
Lau SK, Weiss LM. The Weiss system for evaluating adrenocortical neoplasms: 25 years later. Hum Pathol 2009; 40:757-68. [DOI: 10.1016/j.humpath.2009.03.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 11/29/2022]
|
176
|
Bussey KJ, Demeure MJ. Genomic and expression profiling of adrenocortical carcinoma: application to diagnosis, prognosis and treatment. Future Oncol 2009; 5:641-55. [DOI: 10.2217/fon.09.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is an aggressive endocrine tumor with a poor 5-year survival rate of 10–20%. Current therapy is often ineffective and may be associated with intolerable side effects. Although ACC is extremely rare, recent advances in genomic and expression profiling, coupled with knowledge gained from the study of the inherited syndromes that increase ACC risk, are beginning to bring together a picture of a tumor type dependent on p53, the G2/M cell cycle transition and IGF2 stimulation. Nevertheless, ACC remains a heterogeneous disease. Only recently have sufficient tumors been characterized and results published to permit an exploration of this diversity. Advances in treatment will depend on exploiting those pathways already implicated in ACC, along with those yet to be identified, and testing those treatments in better models of the disease than the three cell lines that currently exist and are widely available to the community.
Collapse
Affiliation(s)
- Kimberly J Bussey
- Clinical Translational Research Division, Translational Genomics Research Institute, 445 N. 5th Street, Phoenix, AZ 85004, USA
| | - Michael J Demeure
- Clinical Translational Research Division, Translational Genomics Research Institute, Director, Endocrine Tumors Center, Scottsdale Healthcare, 10460 N. 92nd St, Suite 200, Scottsdale, AZ 85258, USA
| |
Collapse
|
177
|
Kim AC, Barlaskar FM, Heaton JH, Else T, Kelly VR, Krill KT, Scheys JO, Simon DP, Trovato A, Yang WH, Hammer GD. In search of adrenocortical stem and progenitor cells. Endocr Rev 2009; 30:241-63. [PMID: 19403887 PMCID: PMC2726842 DOI: 10.1210/er.2008-0039] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Scientists have long hypothesized the existence of tissue-specific (somatic) stem cells and have searched for their location in different organs. The theory that adrenocortical organ homeostasis is maintained by undifferentiated stem or progenitor cells can be traced back nearly a century. Similar to other organ systems, it is widely believed that these rare cells of the adrenal cortex remain relatively undifferentiated and quiescent until needed to replenish the organ, at which time they undergo proliferation and terminal differentiation. Historical studies examining cell cycle activation by label retention assays and regenerative potential by organ transplantation experiments suggested that the adrenocortical progenitors reside in the outer periphery of the adrenal gland. Over the past decade, the Hammer laboratory, building on this hypothesis and these observations, has endeavored to understand the mechanisms of adrenocortical development and organ maintenance. In this review, we summarize the current knowledge of adrenal organogenesis. We present evidence for the existence and location of adrenocortical stem/progenitor cells and their potential contribution to adrenocortical carcinomas. Data described herein come primarily from studies conducted in the Hammer laboratory with incorporation of important related studies from other investigators. Together, the work provides a framework for the emerging somatic stem cell field as it relates to the adrenal gland.
Collapse
Affiliation(s)
- Alex C Kim
- Department of Internal Medicine, Division of Metabolism, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Abstract
Adrenocortical carcinoma (ACC) is a rare and heterogeneous malignancy, and most of the diagnostic and therapeutic strategies are not fully established according to criteria of evidence-based medicine. However, recently collaborative efforts (e.g. International Consensus Conference 2003 and networks like the European Network for the Study of Adrenal Tumours (ENSAT)) have significantly advanced the field. This article summarizes current standards in the management of ACC. In patients with suspected ACC a thorough endocrine and imaging work-up is followed by complete (Ro) resection of the tumour by an expert surgeon and initiation of adjuvant mitotane. In advanced disease not amenable to radical resection, cytotoxic drugs will be added to mitotane. The most promising regimens (etoposide, doxorubicin, cisplatin plus mitotane and streptozotocin plus mitotane) are currently compared in an international phase-III trial. Several targeted therapies are under investigation (e.g. IGF-1 inhibitors, sunitinib, sorafenib) and may lead to new treatment options.
Collapse
Affiliation(s)
- Martin Fassnacht
- Dept of Internal Medicine I, Endocrinology and Diabetes Unit, University Hospital, University of Würzburg, Würzburg, Germany.
| | | |
Collapse
|
179
|
Abstract
The study of the clonality of adrenocortical tumours (ACTs) has shown that adrenocortical cancers (ACCs) are of monoclonal origin. Numerous chromosomal alterations have been observed in ACCs, and they are much more frequent than in adrenocortical adenomas. Progress in the genetics of familial syndromes associated with ACTs helped to identify significant somatic molecular alterations in sporadic adult ACCs. Somatic mutations of the tumour suppressor gene TP53 are observed in a third of ACCs. Interestingly, allelic losses (LOH) at the TP53 locus (17p13) are very frequent, observed in more than 85% of ACCs. The insulin-like growth factor II (IGF-II) locus (11p15) is imprinted. IGF-II is over-expressed in 90% of ACCs. Transcriptome studies have identified an IGF-II cluster of genes significantly over-expressed in ACCs. Transcriptome analysis suggests also that the Wnt/beta-catenin signalling pathway is activated in ACT. About a third of ACCs harbours somatic activating mutations of the beta-catenin gene. This recent progress in the molecular genetics of ACC has led to the development of new molecular markers for the diagnosis of malignancy; these might also help to identify prognostic markers of ACC and may ultimately lead to novel therapeutic approaches.
Collapse
Affiliation(s)
- Jérôme Bertherat
- Endocrinology, Metabolism & Cancer Department, INSERM U567, CNRS UMR8104, Institut Cochin, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, INCa-COMETE Centre for Adrenal Cancer, Université Paris-Descartes, Paris, France.
| | | |
Collapse
|
180
|
[Gene profiling and classification of adrenocortical tumors]. ANNALES D'ENDOCRINOLOGIE 2009; 70:186-91. [PMID: 19296923 DOI: 10.1016/j.ando.2009.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
181
|
Cardoso CC, Bornstein SR, Hornsby PJ. New methods for investigating experimental human adrenal tumorigenesis. Mol Cell Endocrinol 2009; 300:175-9. [PMID: 19047010 PMCID: PMC2676229 DOI: 10.1016/j.mce.2008.10.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Revised: 10/28/2008] [Accepted: 10/28/2008] [Indexed: 11/18/2022]
Abstract
Adenomas and nodules of the human adrenal cortex are common, whereas adrenocortical carcinomas are rare. Genes such as IGF2 have been suggested to be important in human adrenocortical tumorigenesis but their role has not been directly investigated. We describe here elements of a system in which hypotheses concerning the molecular basis for the formation of benign and malignant adrenocortical lesions can be experimentally tested. Various viral vectors have been employed in the study of adrenocortical cell biology. Because of the low proliferative rate of primary human adrenocortical (pHAC) cells, a lentiviral system is ideal for transducing these cells with genes that may alter their characteristics or cause them to acquire benign or malignant tumorigenicity. Cultures of pHAC cells were highly infectible with lentiviruses and showed a higher proliferative potential when transduced with a lentivirus encoding IGF2. For tumorigenesis studies of genetically modified adrenocortical cells, we use RAG2(-/-), gammac(-/-) mice. Using this immunodeficient mouse model, we established an orthotopic intra-adrenal cell transplantation technique for adrenocortical cells that should be of value for future studies of the experimental conversion of human adrenocortical cells to a benign or malignant tumorigenic state.
Collapse
Affiliation(s)
- Cibele C Cardoso
- Department of Internal Medicine III, University Medical Center, University of Dresden, Germany
| | | | | |
Collapse
|
182
|
de Reyniès A, Assié G, Rickman DS, Tissier F, Groussin L, René-Corail F, Dousset B, Bertagna X, Clauser E, Bertherat J. Gene expression profiling reveals a new classification of adrenocortical tumors and identifies molecular predictors of malignancy and survival. J Clin Oncol 2009; 27:1108-15. [PMID: 19139432 DOI: 10.1200/jco.2008.18.5678] [Citation(s) in RCA: 282] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Adrenocortical tumors, especially cancers, remain challenging both for their diagnosis and prognosis assessment. The aim of this article is to identify molecular predictors of malignancy and of survival. PATIENTS AND METHODS One hundred fifty-three unilateral adrenocortical tumors were studied by microarray (n = 92) or reverse transcription quantitative polymerase chain reaction (n = 148). A two-gene predictor of malignancy was built using the disease-free survival as the end point in a training cohort (n = 47), then validated in an independent validation cohort (n = 104). The best candidate genes were selected using Cox models, and the best gene combination was validated using the log-rank test. Similarly, for malignant tumors, a two-gene predictor of survival was built using the overall survival as the end point in a training cohort (n = 23), then tested in an independent validation cohort (n = 35). RESULTS Unsupervised clustering analysis discriminated robustly the malignant and benign tumors, and identified two groups of malignant tumors with very different outcome. The combined expression of DLG7 and PINK1 was the best predictor of disease-free survival (log-rank P approximately 10(-12)), could overcome the uncertainties of intermediate pathological Weiss scores, and remained significant after adjustment to the Weiss score (P < 1.3 x 10(-2)). Among the malignant tumors, the combined expression of BUB1B and PINK1 was the best predictor of overall survival (P < 2 x 10(-6)), and remained significant after adjusting for MacFarlane staging (P < .005). CONCLUSION Gene expression analysis unravels two distinct groups of adrenocortical carcinomas. The molecular predictors of malignancy and of survival are reliable and provide valuable independent information in addition to pathology and tumor staging. These original tools should provide important improvements for adrenal tumors management.
Collapse
Affiliation(s)
- Aurélien de Reyniès
- Service des Maladies Endocriniennes et Métaboliques, Hôpital Cochin, 27, rue du Faubourg Saint-Jacques, 75014, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Barlaskar FM, Spalding AC, Heaton JH, Kuick R, Kim AC, Thomas DG, Giordano TJ, Ben-Josef E, Hammer GD. Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. J Clin Endocrinol Metab 2009; 94:204-12. [PMID: 18854392 PMCID: PMC2630877 DOI: 10.1210/jc.2008-1456] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Drug therapy for adrenocortical carcinoma (ACC), a rare and lethal malignancy, is largely empirical and ineffective. New treatments directed at molecular targets critical to the pathophysiology of ACC may prove more efficacious. OBJECTIVE The objective of the study was to profile human adrenal tumors and ACC cell lines to assess activated IGF signaling and determine the efficacy of two IGF receptor (IGF-1R) antagonists alone and in combination with mitotane. EXPERIMENTAL DESIGN ACC cell lines that display or lack activated IGF signaling are used to assess the effects of two IGF-1R antagonists in cultured cells and ACC xenograft tumors. RESULTS Transcriptional profiling data derived from DNA microarray analysis of human adrenal tumors implicate IGF2 as the single highest up-regulated transcript in the vast majority of carcinomas. We show that the majority of ACC cell lines tested display constitutive IGF ligand production and activation of downstream effector pathways. Both IGF-1R antagonists cause significant dose-dependent growth inhibition in ACC cell lines. Furthermore, we observe that mitotane, the first-line adrenolytic drug used in patients with ACC, results in enhanced growth inhibition when used in combination with the IGF-1R antagonists. We next examined the activity of IGF-1R antagonists against ACC xenografts in athymic nude mice. IGF inhibition markedly reduced tumor growth greater than that observed with mitotane treatment, and combination therapy with mitotane significantly enhanced tumor growth suppression. CONCLUSION These findings establish a critical role of IGF signaling in ACC pathophysiology and provide rationale for use of targeted IGF-1R antagonists to treat adrenocortical carcinoma in future clinical trials.
Collapse
Affiliation(s)
- Ferdous M Barlaskar
- Department of Internal Medicine-Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, 1502 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Aubert S. [Adrenal cortex tumors: a lesion continuum?]. Ann Pathol 2008; 28 Spec No 1:S39-41. [PMID: 18984295 DOI: 10.1016/j.annpat.2008.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sébastien Aubert
- Service d'anatomie pathologique, pôle biologie-pathologie, parc Eurasanté, avenue Oscar-Lambret, CHRU, 59037 Lille cedex, France.
| |
Collapse
|
185
|
Tissier F. [Sporadic adrenocortical tumors: genetics and perspectives for the pathologist]. Ann Pathol 2008; 28:409-16. [PMID: 19068395 DOI: 10.1016/j.annpat.2008.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2008] [Indexed: 11/24/2022]
Abstract
Most adrenocortical tumors are benign; adrenocortical carcinomas are rare but their prognosis is poor and few therapeutic options are available. In most adrenocortical tumors, the morphological approach provides enough elements to establish the differential diagnosis between a benign and a malignant tumor but in few cases, it is insufficient. Moreover, morphology is limited for predicting prognosis of adrenocortical carcinomas. These observations led to development of other approaches, in particular immunohistochemical and genetic approaches. The comprehension of the genetic syndromes associated with adrenocortical tumors led to progress in the identification of genetic abnormalities involved in sporadic adrenocortical tumorigenesis. Thus, in sporadic adrenocortical tumorigenesis, IGF-II overexpression and cyclin E overproduction have been associated with 11p15 alterations which are observed in Bethwith-Wiedemann syndrome and TP53 inactivating mutations and 17p13 locus abnormalities which are observed in Li-Fraumeni syndrome. Activation of the Wnt/ss-catenin signaling pathway which is observed in familial adenomatous polyposis has been found in adrenocortical adenomas and carcinomas associated to mutations of CTNNB1, the gene coding ss-catenin, suggesting a central role for this pathway in adrenocortical tumorigenesis. These genetics findings already have had repercussions for patients via the development of molecular markers for diagnosis and prognosis; in the future they should be helpful in the development of new therapeutics.
Collapse
Affiliation(s)
- Fréderique Tissier
- Service d'anatomie pathologique, hôpital Cochin, AP-HP, Paris, France; Faculté de médecine Paris-Descartes, université Paris-Descartes, Paris, France.
| |
Collapse
|
186
|
Waldmann J, Feldmann G, Slater EP, Langer P, Buchholz M, Ramaswamy A, Saeger W, Rothmund M, Fendrich V. Expression of the zinc-finger transcription factor Snail in adrenocortical carcinoma is associated with decreased survival. Br J Cancer 2008; 99:1900-7. [PMID: 19018264 PMCID: PMC2600683 DOI: 10.1038/sj.bjc.6604755] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In this study, we evaluate whether Snail is expressed in adrenocortical cancer (ACC) and if its expression is related to patient outcome. One of the best known functions of the zinc-finger transcription factor Snail is to induce epithelial-to-mesenchymal transition (EMT). Increasing evidence suggests that EMT plays a pivotal role in tumour progression and metastatic spread. Snail and E-cadherin expression were assessed by immunohistochemistry in 26 resected ACCs and real-time quantitative RT–PCR expression analysis was performed. Data were correlated with clinical outcome and in particular with overall patient survival. Seventeen of 26 (65%) ACC tumour samples expressed Snail when assessed by immunohistochemistry. Snail expression was neither detected in normal adrenocortical tissue, nor in benign adrenocortical adenomas. Expression levels were confirmed on the mRNA level by Real-Time–PCR. Survival rates were significantly decreased in Snail-positive tumours compared to Snail-negative tumours: 10 out of 16 vs one out of eight patients succumbed to disease after a median follow up of 14.5 and 28.5 months, respectively (P=0.03). Patients with Snail-expressing ACCs presented in advanced disease (11 out of 12 vs 6 out of 14, P=0.01) and tend to develop distant metastases more frequently than patients with negative staining (7 out of 11 vs two out of eight, P=0.19). In conclusion, we describe for the first time that Snail is expressed in a large subset of ACCs. Furthermore, Snail expression is associated with decreased survival, advanced disease and higher risk of developing distant metastases.
Collapse
Affiliation(s)
- J Waldmann
- Department of Surgery, Philipps-Universität Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Soon PSH, McDonald KL, Robinson BG, Sidhu SB. Molecular markers and the pathogenesis of adrenocortical cancer. Oncologist 2008; 13:548-61. [PMID: 18515740 DOI: 10.1634/theoncologist.2007-0243] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adrenal tumors are common, with an estimated incidence of 7.3% in autopsy cases, while adrenocortical carcinomas (ACCs) are rare, with an estimated prevalence of 4-12 per million population. Because the prognoses for adrenocortical adenomas (ACAs) and ACCs are vastly different, it is important to be able to accurately differentiate the two tumor types. Advancement in the understanding of the pathophysiology of ACCs is essential for the development of more sensitive means of diagnosis and treatment, resulting in better clinical outcome. Adrenocortical tumors (ACTs) occur as a component of several hereditary tumor syndromes, which include the Li-Fraumeni syndrome, Beckwith-Wiedemann syndrome, multiple endocrine neoplasia 1, Carney complex, and congenital adrenal hyperplasia. The genes involved in these syndromes have also been shown to play a role in the pathogenesis of sporadic ACTs. The adrenocorticotropic hormone-cAMP-protein kinase A and Wnt pathways are also implicated in adrenocortical tumorigenesis. The aim of this review is to summarize the current knowledge on the molecular mechanisms involved in adrenocortical tumorigenesis, including results of comparative genomic hybridization, loss of heterozygosity, and microarray gene-expression profiling studies.
Collapse
Affiliation(s)
- Patsy S H Soon
- Cancer Genetics, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
188
|
Hironobu S, Takashi S, Yasuhiro N. Discerning malignancy in resected adrenocotical tumors. ACTA ACUST UNITED AC 2008; 2:1095-105. [PMID: 23496421 DOI: 10.1517/17530059.2.10.1095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The number of adrenocortical tumors discovered incidentally has recently increased owing to the advent of imaging techniques. The most important diagnostic point in evaluation of resected specimens of these tumors is to discern malignancy to determine postoperative management of the patients. OBJECTIVE/METHODS To determine what the effective methods of discerning malignancy in resected specimens of adrenocortical tumors are at this juncture. To provide relevant and practical information pertinent to those involved in the management of patients with adrenocortical tumors. CONCLUSION Careful macroscopic evaluation, including the selection of the specimens submitted and application of the criteria of Weiss for histological diagnosis, is still considered the 'gold standard' for diagnosis of adrenocortical carcinoma. However, it is important to recognize its limitations in the diagnosis of adrenocortical oncocytoma and pediatric adrenocortical tumors. The auxiliary methods that may be of clinical relevance at this juncture include the analysis of the Ki67/MIB-1 labeling index and IGF-II expression in the tumors.
Collapse
Affiliation(s)
- Sasano Hironobu
- Tohoku University School of Medicine, Department of Pathology, 2-1 Seiryou-machi, Aoba-ku, Sendai, 980-8575, Japan +81 22 717 7450 ; +81 22 273 5976 ;
| | | | | |
Collapse
|
189
|
Rosati R, Cerrato F, Doghman M, Pianovski MAD, Parise GA, Custódio G, Zambetti GP, Ribeiro RC, Riccio A, Figueiredo BC, Lalli E. High frequency of loss of heterozygosity at 11p15 and IGF2 overexpression are not related to clinical outcome in childhood adrenocortical tumors positive for the R337H TP53 mutation. ACTA ACUST UNITED AC 2008; 186:19-24. [PMID: 18786438 DOI: 10.1016/j.cancergencyto.2008.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2008] [Revised: 05/19/2008] [Accepted: 05/21/2008] [Indexed: 11/16/2022]
Abstract
A germline TP53 R337H mutation is present in childhood adrenocortical tumors (ACT) from southern Brazil. Other genetic alterations are also frequently found in these tumors. This study was designed to assess whether alterations of the 11p15 region exist in childhood ACT, accounting for IGF2 overexpression in these tumors, and how they are related to clinical outcome. Tumor DNA of 12 children with ACT (4 adenomas and 8 carcinomas) and from the blood of their parents was analyzed. All patients showed 11p15 loss of heterozygosity (LOH) in the tumor. In contrast to the single case of paternal LOH, IGF2 was overexpressed in tumors with maternal allele loss. Our data show that 11p15 LOH is a widespread finding in childhood ACT not related with malignancy, contrary to adult ACT. Alterations in the expression of other genes in the same region (e.g., CDKN1C) may contribute to ACT tumorigenesis.
Collapse
Affiliation(s)
- Roberto Rosati
- Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Av. Silva Jardim, Paraná, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Almeida MQ, Fragoso MCBV, Lotfi CFP, Santos MG, Nishi MY, Costa MHS, Lerario AM, Maciel CC, Mattos GE, Jorge AAL, Mendonca BB, Latronico AC. Expression of insulin-like growth factor-II and its receptor in pediatric and adult adrenocortical tumors. J Clin Endocrinol Metab 2008; 93:3524-31. [PMID: 18611974 DOI: 10.1210/jc.2008-0065] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Adrenocortical tumors are heterogeneous neoplasms with incompletely understood pathogenesis. IGF-II overexpression has been consistently demonstrated in adult adrenocortical carcinomas. OBJECTIVES The objective of the study was to analyze expression of IGF-II and its receptor (IGF-IR) in pediatric and adult adrenocortical tumors and the effects of a selective IGF-IR kinase inhibitor (NVP-AEW541) on adrenocortical tumor cells. PATIENTS Fifty-seven adrenocortical tumors (37 adenomas and 20 carcinomas) from 23 children and 34 adults were studied. METHODS Gene expression was determined by quantitative real-time PCR. Cell proliferation and apoptosis were analyzed in NCI H295 cells and a new cell line established from a pediatric adrenocortical adenoma. RESULTS IGF-II transcripts were overexpressed in both pediatric adrenocortical carcinomas and adenomas. Otherwise, IGF-II was mainly overexpressed in adult adrenocortical carcinomas (270.5 +/- 130.2 vs. 16.1 +/- 13.3; P = 0.0001). IGF-IR expression was significantly higher in pediatric adrenocortical carcinomas than adenomas (9.1 +/- 3.1 vs. 2.6 +/- 0.3; P = 0.0001), whereas its expression was similar in adult adrenocortical carcinomas and adenomas. IGF-IR expression was a predictor of metastases in pediatric adrenocortical tumors in univariate analysis (hazard ratio 1.84; 95% confidence interval 1.28-2.66; P = 0.01). Furthermore, NVP-AEW541 blocked cell proliferation in a dose- and time-dependent manner in both cell lines through a significant increase of apoptosis. CONCLUSION IGF-IR overexpression was a biomarker of pediatric adrenocortical carcinomas. Additionally, a selective IGF-IR kinase inhibitor had antitumor effects in adult and pediatric adrenocortical tumor cell lines, suggesting that IGF-IR inhibitors represent a promising therapy for human adrenocortical carcinoma.
Collapse
Affiliation(s)
- Madson Q Almeida
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM-42 da Disciplina de Endocrinologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, SP, Brasil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Tadjine M, Lampron A, Ouadi L, Horvath A, Stratakis CA, Bourdeau I. Detection of somatic beta-catenin mutations in primary pigmented nodular adrenocortical disease (PPNAD). Clin Endocrinol (Oxf) 2008; 69:367-73. [PMID: 18419788 PMCID: PMC3138207 DOI: 10.1111/j.1365-2265.2008.03273.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Primary pigmented nodular adrenocortical disease (PPNAD) leads to Cushing syndrome (CS) and is often associated with Carney complex (CNC). Genetic alterations of the type 1-alpha regulatory subunit of cAMP-dependent protein kinase A (PRKAR1A) and phosphodiesterase 11A4 (PDE11A) genes have been found in PPNAD. Recent studies have demonstrated that beta-catenin mutations are frequent in adrenocortical adenomas and carcinomas and that the Wnt-signalling pathway is involved in PPNAD tumorigenesis. We hypothesized that adrenocortical adenomas that form in the context of PPNAD may harbour beta-catenin mutations. METHODS We studied 18 patients with CS secondary to PPNAD who were screened for germline PRKAR1A and PDE11A mutations. Tumor DNA was extracted from pigmented adrenocortical adenoma and nodular adrenal hyperplasia. Mutation analysis of exons 3 and 5 of beta-catenin was performed using polymerase chain reaction and direct sequencing. Sections from formalin-fixed, paraffin-embedded tumour samples were studied by immunohistochemistry with an antibody against beta-catenin. RESULTS Nine patients were carrying germline PRKAR1A mutations and one patient had a PDE11A mutation. We found somatic beta-catenin mutations in 2 of 18 patients (11%). In both cases, the mutations occurred in relatively large adenomas that had formed in the background of PPNAD. Tumor DNA analysis revealed a heterozygous ACC-to-GCC missense mutation in codon 41 (T41A) and a TCT-to-CCT missense mutation in codon 45 (S45P) of exon 3 of the beta-catenin gene that was confirmed at the cDNA level. There were no alterations in the DNA of PPNAD-adjacent tissues and lymphocytes from the patients, indicating somatic events. Immunohistochemistry showed nuclear accumulation of beta-catenin in more than 90% of cells in adenomatous tissue whereas no nuclear immunoreactivity was detected in adjacent PPNAD nodular cells. Nuclear translocation of beta-catenin protein in the PPNAD adenoma suggests activation of the Wnt-beta-catenin pathway in PPNAD. CONCLUSIONS We report, for the first time, beta-catenin mutations in adenomas associated with PPNAD, further implicating Wnt-beta-catenin signalling in tumorigenesis linked to bilateral adrenal hyperplasias.
Collapse
Affiliation(s)
- Mimi Tadjine
- Division of Endocrinology, Department of Medicine, Centre hospitalier de l’Université de Montréal (CHUM) Hôtel-Dieu, Montreal, QC, Canada
| | - Antoine Lampron
- Division of Endocrinology, Department of Medicine, Centre hospitalier de l’Université de Montréal (CHUM) Hôtel-Dieu, Montreal, QC, Canada
| | - Lydia Ouadi
- Division of Endocrinology, Department of Medicine, Centre hospitalier de l’Université de Montréal (CHUM) Hôtel-Dieu, Montreal, QC, Canada
| | - Anelia Horvath
- Pediatric Endocrinology Inter-institute Training Program, NICHD, NIH, Bethesda, MD, USA
| | | | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine, Centre hospitalier de l’Université de Montréal (CHUM) Hôtel-Dieu, Montreal, QC, Canada
| |
Collapse
|
192
|
Abstract
CONTEXT In surgical pathology practice adrenal cortical tumors are rare. However, in autopsy series adrenal cortical nodules are found frequently. These are now being identified more commonly in life when the abdomen is scanned for other disease. It is important to differentiate between benign and malignant lesions as adrenal cortical carcinoma is an aggressive tumor. Molecular genetic investigations are providing new information on both pathogenesis of adrenal tumors and basic adrenal development and physiology. OBJECTIVE To provide an overview of current knowledge on adrenal cortical development and structure that informs our understanding of genetic diseases of the adrenal cortex and adrenal cortical tumors. DATA SOURCES Literature review using PubMed via the Endnote bibliography tool. CONCLUSIONS The understanding of basic developmental and physiologic processes permits a better understanding of diseases of the adrenal cortex. The information coming from investigation of the molecular pathology of adrenal cortical tumors is beginning to provide additional tests for the assessment of malignant potential in diagnosis but the mainstay remains traditional histologic analysis.
Collapse
Affiliation(s)
- Anne Marie McNicol
- Pathology Department, University of Glasgow, Royal Infirmary, Glasgow, United Kingdom.
| |
Collapse
|
193
|
Transcriptome analysis of endocrine tumors: clinical perspectives. ANNALES D'ENDOCRINOLOGIE 2008; 69:130-4. [PMID: 18423557 DOI: 10.1016/j.ando.2008.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
There is considerable interest in the application of DNA microarrays to the pathologic evaluation of endocrine neoplasms. Improvements in tumor classification and prognostication, prediction of response to therapy, and comprehensive assessment of tumoral hormone production represent the major anticipated benefits. Here, some of the microarray studies that support the clinical use of transcriptome profiling for endocrine tumors are reviewed. In addition, some of the barriers to clinical implementation are discussed.
Collapse
|
194
|
Ito Y, Koessler T, Ibrahim AEK, Rai S, Vowler SL, Abu-Amero S, Silva AL, Maia AT, Huddleston JE, Uribe-Lewis S, Woodfine K, Jagodic M, Nativio R, Dunning A, Moore G, Klenova E, Bingham S, Pharoah PDP, Brenton JD, Beck S, Sandhu MS, Murrell A. Somatically acquired hypomethylation of IGF2 in breast and colorectal cancer. Hum Mol Genet 2008; 17:2633-43. [PMID: 18541649 PMCID: PMC2515372 DOI: 10.1093/hmg/ddn163] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The imprinted insulin-like growth factor 2 (IGF2) gene is expressed predominantly from the paternal allele. Loss of imprinting (LOI) associated with hypomethylation at the promoter proximal sequence (DMR0) of the IGF2 gene was proposed as a predisposing constitutive risk biomarker for colorectal cancer. We used pyrosequencing to assess whether IGF2 DMR0 methylation is either present constitutively prior to cancer or whether it is acquired tissue-specifically after the onset of cancer. DNA samples from tumour tissues and matched non-tumour tissues from 22 breast and 42 colorectal cancer patients as well as peripheral blood samples obtained from colorectal cancer patients [SEARCH (n=case 192, controls 96)], breast cancer patients [ABC (n=case 364, controls 96)] and the European Prospective Investigation of Cancer [EPIC-Norfolk (n=breast 228, colorectal 225, controls 895)] were analysed. The EPIC samples were collected 2–5 years prior to diagnosis of breast or colorectal cancer. IGF2 DMR0 methylation levels in tumours were lower than matched non-tumour tissue. Hypomethylation of DMR0 was detected in breast (33%) and colorectal (80%) tumour tissues with a higher frequency than LOI indicating that methylation levels are a better indicator of cancer than LOI. In the EPIC population, the prevalence of IGF2 DMR0 hypomethylation was 9.5% and this correlated with increased age not cancer risk. Thus, IGF2 DMR0 hypomethylation occurs as an acquired tissue-specific somatic event rather than a constitutive innate epimutation. These results indicate that IGF2 DMR0 hypomethylation has diagnostic potential for colon cancer rather than value as a surrogate biomarker for constitutive LOI.
Collapse
Affiliation(s)
- Yoko Ito
- Department of Oncology, University of Cambridge, CRUK Cambridge Research Institute, Li Ka- Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Le cancer de la surrénale. Comment optimiser la prise en charge d’un cancer rare? UPC Inca, COMETE. ONCOLOGIE 2008. [DOI: 10.1007/s10269-008-0899-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
196
|
Stratakis CA, Horvath A. How the new tools to analyze the human genome are opening new perspectives: the use of gene expression in investigations of the adrenal cortex. ANNALES D'ENDOCRINOLOGIE 2008; 69:123-9. [PMID: 18423555 DOI: 10.1016/j.ando.2008.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
With the promise of state-of-the-art molecular technologies and the tools provided by the human genome project, a number of investigators are trying to identify molecular targets of adrenocortical tumorigenesis. One path in this endeavor was the identification by positional cloning of genes that are mutated in rare adrenocortical tumors. The subject of this article is an update of the results of experiments in the second path that was followed by us and others: that of using genome-wide expression analysis of adrenocortical cells in normal and various disease states. Transcriptomic analysis is a rapidly evolving technology; this article summarizes some data on the adrenal cortex and points out how these new technologies can be used in the identification of important genes and molecular pathways in both normal and diseased adrenal cortex.
Collapse
Affiliation(s)
- C A Stratakis
- Section on Endocrinology, Genetics, Program on Developmental Endocrinology & Genetics, National Institute of Child Health and Human Development, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1862, USA.
| | | |
Collapse
|
197
|
Fernandez-Ranvier GG, Weng J, Yeh RF, Shibru D, Khafnashar E, Chung KW, Hwang J, Duh QY, Clark OH, Kebebew E. Candidate Diagnostic Markers and Tumor Suppressor Genes for Adrenocortical Carcinoma by Expression Profile of Genes on Chromosome 11q13. World J Surg 2008; 32:873-81. [PMID: 18324346 DOI: 10.1007/s00268-008-9521-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gustavo G Fernandez-Ranvier
- Department of Surgery, University of California, San Francisco, School of Medicine, UCSF/Mt. Zion Medical Center, 1600 Divisadero Street, San Francisco, CA 94143-1674, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Abstract
The network regulating human adrenal development is complex. Studies of patients with adrenal insufficiency due to gene mutations established a central role for transcription factors GLI3, SF1 and DAX1 in the initial steps of adrenal formation. Adrenal differentiation seems to depend on adrenocorticotropic hormone (ACTH) stimulation and signalling, including biosynthesis and action of POMC, PC1, TPIT, MC2R, MRAP and ALADIN, all of which cause adrenocortical hypoplasia when mutated in humans. Studies of knockout mice revealed many more factors involved in adrenal development; however, in contrast to rodents, in humans several of those factors had no adrenal phenotype when mutated (e.g. WT1, WNT4) or, alternatively, human mutations have not (yet) been identified. Tissue profiling of fetal and adult adrenals suggested 69 genes involved in adrenal development. Among them were genes coding for steroidogenic enzymes, transcription and growth factors, signalling molecules, regulators of cell cycle and angiogenesis, and extracellular matrix proteins; however, the exact role of most of them remains to be elucidated.
Collapse
Affiliation(s)
- Petra Kempná
- Pediatric Endocrinology and Diabetology, University Children's Hospital, University of Berne, Freiburgstrasse 15, 3010 Bern, Switzerland
| | | |
Collapse
|
199
|
Abstract
The adrenal gland is not a common specimen in surgical pathology practice as, until recently, adrenal tumors were recognized in life only if associated with hypersecretion of hormones or evidence of malignancy. However, adrenal nodules are not uncommon at autopsy, and the number of these found in life is now increasing as they are identified when the abdomen is scanned for the investigation of other diseases using computed tomography or magnetic resonance imaging. It is therefore becoming increasingly important for the surgical pathologist to be aware of the range of pathology in the gland and to understand how to approach the specimens. This short review will deal with lesions of the adrenal cortex.
Collapse
Affiliation(s)
- Anne Marie McNicol
- Molecular and Cellular Pathology, School of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
200
|
Affiliation(s)
- Ferdous M Barlaskar
- Cellular & Molecular Biology Graduate Program, University of Michigan, BSRB 1502, Ann Arbor, MI 48109-2200, USA
| | | |
Collapse
|