1
|
Wakefield CB, Sadow PM, Hornick JL, Fletcher CDM, Barletta JA, Anderson WJ. PEComa of the Adrenal Gland: A Clinicopathologic Series of 7 Cases. Am J Surg Pathol 2023; 47:1316-1324. [PMID: 37545320 DOI: 10.1097/pas.0000000000002097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
PEComas are a family of mesenchymal neoplasms composed of histologically distinctive perivascular epithelioid cells which demonstrate myomelanocytic differentiation. PEComas of the adrenal gland are very rare and can represent a considerable diagnostic challenge given their morphologic overlap with more common adrenal cortical neoplasms. We present the clinicopathologic features of 7 primary adrenal PEComas. The cohort comprised 5 male and 2 female patients with a median age of 63 years (range: 31 to 71 y). One patient had Birt-Hogg-Dubé syndrome and another had Lynch syndrome; however, none had a history of tuberous sclerosis complex. Histologically, tumors showed nested and/or sheet-like growth and epithelioid cytomorphology with pale-to-eosinophilic granular cytoplasm. Two tumors had an admixed spindle cell component. There was a median of 4 mitoses per 10 HPFs (range: 0 to 8). Necrosis was present in 4 tumors and lymphovascular invasion in 1. Four tumors were classified as malignant. By immunohistochemistry, tumors were positive for HMB-45 (3/7), MITF (3/3), Melan-A (3/7), smooth muscle actin (5/7), desmin (5/7), and caldesmon (1/1). Two tumors were positive for TFE3 (2/4). Inhibin and SF1 were negative in all tumors assessed (0/6). Of 3 patients with available clinical follow-up information, 1 patient developed locally recurrent and metastatic disease (at 18 mo) and was alive with persistent disease at the last follow-up. Two patients had no recurrent or metastatic disease at the last follow-up (60 and 25 mo). Although PEComas of the adrenal gland are rare, pathologists need to be alert to this entity in the differential diagnosis of primary adrenocortical neoplasms. In suspected cases, the judicious use of melanocytic and smooth muscle markers, in addition to TFE3 and markers of adrenocortical differentiation (such as SF1 and inhibin) can assist in diagnosis. As in PEComas arising at other visceral sites, an association with tuberous sclerosis complex seems to be uncommon.
Collapse
Affiliation(s)
- Craig B Wakefield
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School
| | | | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School
| | - William J Anderson
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School
| |
Collapse
|
2
|
Plaska SW, Liu CJ, Lim JS, Rege J, Bick NR, Lerario AM, Hammer GD, Giordano TJ, Else T, Tomlins SA, Rainey WE, Udager AM. Targeted RNAseq of Formalin-Fixed Paraffin-Embedded Tissue to Differentiate Among Benign and Malignant Adrenal Cortical Tumors. Horm Metab Res 2020; 52:607-613. [PMID: 32791542 PMCID: PMC7880170 DOI: 10.1055/a-1212-8803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lack of routine fresh or frozen tissue is a barrier to widespread transcriptomic analysis of adrenal cortical tumors and an impediment to translational research in endocrinology and endocrine oncology. Our group has previously pioneered the use of targeted amplicon-based next-generation sequencing for archival formalin-fixed paraffin-embedded (FFPE) adrenal tissue specimens to characterize the spectrum of somatic mutations in various forms of primary aldosteronism. Herein, we developed and validated a novel 194-amplicon targeted next-generation RNA sequencing (RNAseq) assay for transcriptomic analysis of adrenal tumors using clinical-grade FFPE specimens. Targeted RNAseq-derived expression values for 27 adrenal cortical tumors, including aldosterone-producing adenomas (APA; n=8), cortisol-producing adenomas (CPA; n=11), and adrenal cortical carcinomas (ACC; n=8), highlighted known differentially-expressed genes (DEGs; i. e., CYP11B2, IGF2, etc.) and tumor type-specific transcriptional modules (i. e., high cell cycle/proliferation transcript expression in ACC, etc.), and a subset of DEGs was validated orthogonally using quantitative reverse transcription PCR (qRT-PCR). Finally, unsupervised hierarchical clustering using a subset of high-confidence DEGs revealed three discrete clusters representing APA, CPA, and ACC tumors with corresponding unique gene expression signatures, suggesting potential clinical utility for a transcriptomic-based approach to tumor classification. Overall, these data support the use of targeted amplicon-based RNAseq for comprehensive transcriptomic profiling of archival FFPE adrenal tumor material and indicate that this approach may facilitate important translational research opportunities for the study of these tumors.
Collapse
Affiliation(s)
- Samuel W. Plaska
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Chia-Jen Liu
- Michigan Center for Translational Pathology, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jung Soo Lim
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nolan R. Bick
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Antonio M. Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrine, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gary D. Hammer
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Metabolism, Endocrine, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas J. Giordano
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Metabolism, Endocrine, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Tobias Else
- Department of Internal Medicine, Division of Metabolism, Endocrine, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Scott A. Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Metabolism, Endocrine, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Co-corresponding authors: Aaron M. Udager, MD, PhD, Department of Pathology, University of Michigan Medical School, 3308 Rogel Cancer Center, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109, UDA, , Phone: (734) 232-6399, Fax: (734) 763-4095, William E. Rainey, PhD, Department of Molecular and Integrative Physiology, University of Michigan Medical School, 2558 Medical Science Research Building II, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA, , Phone: (734) 764-7514, Fax: (734) 936-8813
| | - Aaron M. Udager
- Michigan Center for Translational Pathology, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Co-corresponding authors: Aaron M. Udager, MD, PhD, Department of Pathology, University of Michigan Medical School, 3308 Rogel Cancer Center, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109, UDA, , Phone: (734) 232-6399, Fax: (734) 763-4095, William E. Rainey, PhD, Department of Molecular and Integrative Physiology, University of Michigan Medical School, 2558 Medical Science Research Building II, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA, , Phone: (734) 764-7514, Fax: (734) 936-8813
| |
Collapse
|
3
|
Zlatibor L, Paunovic I, Zivaljevic V, Dundjerovic D, Tatic S, Djukic V. Prognostic significance of immunohistochemical markers in adrenocortical carcinoma. Acta Chir Belg 2020; 120:23-29. [PMID: 30499377 DOI: 10.1080/00015458.2018.1543822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background: To present basic demographic and clinical characteristics of patients with adrenocortical carcinoma (ACC), to determine the overall survival rate and to analyze the results of immunohistochemical staining and its correlation with the length of survival.Material and methods: The study was conducted during the period between 1996 and 2010 and included 30 patients with ACC. Immunohistochemical staining (MMP9, melan A, inhibin, caltretinin, D2-40, synaptophysin and Ki-67) was performed.Results: ACC was diagnosed in 19 females and 11 men (1.7:1). The average age was 50.1 years. The median tumor size was 10 cm, the median weight 400 g. Majority of subjects had positive immunohistochemical staining for the markers of interest. Patients with any negative staining had shorter cancer-specific survival than ones with positive staining. According to the log-rank test results as well as according to the results of the univariate Cox analysis, negative staining for inhibin, D2-40 and synaptophysin and Ki-67 expression ≥7% were associated with poorer prognosis.Conclusions: The results of our study suggest that the absence of staining for some immunohistochemical markers and increased expression of Ki-67 are associated with a poorer prognosis and shorter survival of patients with ACC. Immunohistochemical markers may serve as a prognostic factor for ACC.
Collapse
Affiliation(s)
- Loncar Zlatibor
- 1Emergency Centre, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivan Paunovic
- 2Centre for Endocrine Surgery, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladan Zivaljevic
- 2Centre for Endocrine Surgery, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dusko Dundjerovic
- 3Faculty of Medicine, Institute of Pathology, University of Belgrade, Belgrade, Serbia
| | - Svetislav Tatic
- 3Faculty of Medicine, Institute of Pathology, University of Belgrade, Belgrade, Serbia
| | - Vladimir Djukic
- 1Emergency Centre, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
6
|
Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J, Willscher E, Walz MK, Worm K, Schmid KW. Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. J Clin Pathol 2011; 64:529-35. [PMID: 21471143 PMCID: PMC3099361 DOI: 10.1136/jcp.2010.085621] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND For the clinical management of adrenocortical neoplasms it is crucial to correctly distinguish between benign and malignant tumours. Even histomorphologically based scoring systems do not allow precise separation in single lesions, thus novel parameters are desired which offer a more accurate differentiation. The tremendous potential of microRNAs (miRNAs) as diagnostic biomarkers in surgical pathology has recently been shown in a broad variety of tumours. METHODS In order to elucidate the diagnostic impact of miRNA expression in adrenocortical neoplasms, a cohort of 20 adrenocortical specimens including normal adrenal tissue (n=4), adrenocortical adenomas (ACAs) (n=9), adrenocortical carcinomas (ACCs) (n=4) and metastases (n=3) was analysed using TaqMan low density arrays to identify specific miRNA profiles in order to distinguish between benign and malignant adrenocortical lesions. Results were validated in a validation cohort (n=16). RESULTS Concerning the differential diagnosis of ACAs and ACCs, 159 out of 667 miRNAs were up- and 89 were down-regulated in ACAs. Using real-time PCR analysis of three of the most significantly expressed single key miRNAs allowed separation of ACAs from ACCs. ACCs exhibited significantly lower levels of miR-139-3p (up to 8.49-fold, p<0.001), miR-675 (up to 23.25-fold, p<0.001) and miR-335 (up to 5.25-fold, p<0.001). A validation cohort of 16 specimen with known Weiss score showed up-regulation of miR-335 and miR-675 in the majority of cases with probable malignant course, although overlapping values exist. CONCLUSION miRNA profiling of miR-675 and miR-335 helps in discriminating ACCs from ACAs. miRNA analysis may indicate malignant behaviour in cases with indeterminate malignant potential.
Collapse
Affiliation(s)
- K J Schmitz
- Institute of Pathology and Neuropathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
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: 16.9] [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
|
13
|
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
|