1
|
Ma Y, Field NR, Xie T, Briscas S, Kokinogoulis EG, Skipper TS, Alghalayini A, Sarker FA, Tran N, Bowden NA, Dickson KA, Marsh DJ. Aberrant SWI/SNF Complex Members Are Predominant in Rare Ovarian Malignancies-Therapeutic Vulnerabilities in Treatment-Resistant Subtypes. Cancers (Basel) 2024; 16:3068. [PMID: 39272926 PMCID: PMC11393890 DOI: 10.3390/cancers16173068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
SWI/SNF (SWItch/Sucrose Non-Fermentable) is the most frequently mutated chromatin-remodelling complex in human malignancy, with over 20% of tumours having a mutation in a SWI/SNF complex member. Mutations in specific SWI/SNF complex members are characteristic of rare chemoresistant ovarian cancer histopathological subtypes. Somatic mutations in ARID1A, encoding one of the mutually exclusive DNA-binding subunits of SWI/SNF, occur in 42-67% of ovarian clear cell carcinomas (OCCC). The concomitant somatic or germline mutation and epigenetic silencing of the mutually exclusive ATPase subunits SMARCA4 and SMARCA2, respectively, occurs in Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT), with SMARCA4 mutation reported in 69-100% of SCCOHT cases and SMARCA2 silencing seen 86-100% of the time. Somatic ARID1A mutations also occur in endometrioid ovarian cancer (EnOC), as well as in the chronic benign condition endometriosis, possibly as precursors to the development of the endometriosis-associated cancers OCCC and EnOC. Mutation of the ARID1A paralogue ARID1B can also occur in both OCCC and SCCOHT. Mutations in other SWI/SNF complex members, including SMARCA2, SMARCB1 and SMARCC1, occur rarely in either OCCC or SCCOHT. Abrogated SWI/SNF raises opportunities for pharmacological inhibition, including the use of DNA damage repair inhibitors, kinase and epigenetic inhibitors, as well as immune checkpoint blockade.
Collapse
Affiliation(s)
- Yue Ma
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Natisha R Field
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Tao Xie
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sarina Briscas
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Emily G Kokinogoulis
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Tali S Skipper
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Amani Alghalayini
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Farhana A Sarker
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Nham Tran
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Nikola A Bowden
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, Newcastle, NSW 2289, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2289, Australia
| | - Kristie-Ann Dickson
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Deborah J Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
2
|
Morgan JE, Jaferi N, Shonibare Z, Huang GS. ARID1A in Gynecologic Precancers and Cancers. Reprod Sci 2024; 31:2150-2162. [PMID: 38740655 DOI: 10.1007/s43032-024-01585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
The highest frequency of genetic alterations in the tumor suppressor ARID1A occurs in malignancies of the female reproductive tract. The prevalence of ARID1A alterations in gynecologic precancers and cancers is summarized from the literature, and the putative mechanisms of tumor suppressive action examined both in benign/precursor lesions including endometriosis and atypical hyperplasia and in malignancies of the ovary, uterus, cervix and vagina. ARID1A alterations in gynecologic cancers are usually loss-of-function mutations, resulting in diminished or absent protein expression. ARID1A deficiency results in pleiotropic downstream effects related not only to its role in transcriptional regulation as a SWI/SNF complex subunit, but also related to the functions of ARID1A in DNA replication and repair, immune modulation, cell cycle progression, endoplasmic reticulum (ER) stress and oxidative stress. The most promising actionable signaling pathway interactions and therapeutic vulnerabilities of ARID1A mutated cancers are presented with a critical review of the currently available experimental and clinical evidence. The role of ARID1A in response to chemotherapeutic agents, radiation therapy and immunotherapy is also addressed. In summary, the multi-faceted role of ARID1A mutation in precancer and cancer is examined through a clinical lens focused on development of novel preventive and therapeutic interventions for gynecological cancers.
Collapse
Affiliation(s)
- Jaida E Morgan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Nishah Jaferi
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Zainab Shonibare
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA.
- Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology, Yale School of Medicine, Yale Cancer Center, Yale University, PO Box 208063, New Haven, CT, 06520-8063, USA.
| |
Collapse
|
3
|
O'Cearbhaill RE, Miller A, Soslow RA, Lankes HA, DeLair D, Segura S, Chavan S, Zamarin D, DeBernardo R, Moore K, Moroney J, Shahin M, Thaker PH, Wahner-Hendrickson AE, Aghajanian C. A phase 2 study of dasatinib in recurrent clear cell carcinoma of the ovary, fallopian tube, peritoneum or endometrium: NRG oncology/gynecologic oncology group study 0283. Gynecol Oncol 2023; 176:16-24. [PMID: 37418832 PMCID: PMC10529107 DOI: 10.1016/j.ygyno.2023.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
OBJECTIVE Gynecologic cancers are traditionally managed according to their presumed site of origin, without regard to the underlying histologic subtype. Clear cell histology is associated with chemotherapy refractoriness and poor survival. Mutations in SWI/SNF chromatin remodeling complex member ARID1A, which encodes for BAF250a protein, are common in clear cell and endometriosis-associated endometrioid carcinomas. High-throughput cell-based drug screening predicted activity of dasatinib, a tyrosine kinase inhibitor, in ARID1A-mutant clear cell carcinoma. METHODS We conducted a phase 2 clinical trial of dasatinib 140 mg once daily by mouth in patients with recurrent or persistent ovarian and endometrial clear cell carcinoma. Patients with measurable disease were enrolled and then assigned to biomarker-defined populations based on BAF250a immunohistochemistry. The translational endpoints included broad next-generation sequencing to assess concordance of protein expression and treatment outcomes. RESULTS Twenty-eight patients, 15 of whom had tumors with retained BAF250a and 13 with loss of BAF250a were evaluable for treatment response and safety. The most common grade 3 adverse events were anemia, fatigue, dyspnea, hyponatremia, pleural effusion, and vomiting. One patient had a partial response, eight (28%) had stable disease, and 15 (53.6%) had disease progression. Twenty-three patients had next-generation sequencing results; 13 had a pathogenic ARID1A alteration. PIK3CA mutations were more prevalent in ARID1A-mutant tumors, while TP53 mutations were more prevalent in ARID1A wild-type tumors. CONCLUSIONS Dasatinib was not an effective single-agent treatment for recurrent or persistent ovarian and endometrial clear cell carcinoma. Studies are urgently needed for this rare gynecologic subtype.
Collapse
Affiliation(s)
| | - Austin Miller
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America.
| | - Robert A Soslow
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| | - Heather A Lankes
- NRG Oncology, Operations Center-Philadelphia East, Philadelphia, PA, United States of America; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America.
| | - Deborah DeLair
- Northwell Health, Greenvale, New York, NY, United States of America.
| | - Sheila Segura
- Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - Shweta Chavan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| | - Dmitriy Zamarin
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| | | | - Kathleen Moore
- University of Oklahoma, Oklahoma City, OK, United States of America.
| | - John Moroney
- University of Chicago, Chicago, IL, United States of America.
| | - Mark Shahin
- Abington Memorial Hospital, Willow Grove, PA, United States of America.
| | - Premal H Thaker
- Washington University, St. Louis, MO, United States of America.
| | | | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| |
Collapse
|
4
|
Hinsberger M, Becker-Kettern J, Jürgens-Wemheuer WM, Oertel J, Schulz-Schaeffer WJ. Development of an Enzyme-Linked Immunosorbent Assay (ELISA) for the Quantification of ARID1A in Tissue Lysates. Cancers (Basel) 2023; 15:4096. [PMID: 37627124 PMCID: PMC10452747 DOI: 10.3390/cancers15164096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
ARID1A is a subunit of the mammalian SWI/SNF complex, which is thought to regulate gene expression through restructuring chromatin structures. Its gene ARID1A is frequently mutated and ARID1A levels are lowered in several human cancers, especially gynecologic ones. A functional ARID1A loss may have prognostic or predictive value in terms of therapeutic strategies but has not been proposed based on a quantitative method. Hardly any literature is available on ARID1A levels in tumor samples. We developed an indirect enzyme-linked immunosorbent assay (ELISA) for ARID1A based on the current EMA and FDA criteria. We demonstrated that our ELISA provides the objective, accurate, and precise quantification of ARID1A concentrations in recombinant protein solutions, cell culture standards, and tissue lysates of tumors. A standard curve analysis yielded a 'goodness of fit' of R2 = 0.99. Standards measured on several plates and days achieved an inter-assay accuracy of 90.26% and an inter-assay precision with a coefficient of variation of 4.53%. When tumor lysates were prepared and measured multiple times, our method had an inter-assay precision with a coefficient of variation of 11.78%. We believe that our suggested method ensures a high reproducibility and can be used for a high sample throughput to determine the ARID1A concentration in different tumor entities. The application of our ELISA on various tumor and control tissues will allow us to explore whether quantitative ARID1A measurements in tumor samples are of predictive value.
Collapse
Affiliation(s)
- Manuel Hinsberger
- Institute for Neuropathology, Medical Faculty, Saarland University, Building 90.3, 66421 Homburg, Saar, Germany (J.B.-K.); (W.M.J.-W.)
| | - Julia Becker-Kettern
- Institute for Neuropathology, Medical Faculty, Saarland University, Building 90.3, 66421 Homburg, Saar, Germany (J.B.-K.); (W.M.J.-W.)
| | - Wiebke M. Jürgens-Wemheuer
- Institute for Neuropathology, Medical Faculty, Saarland University, Building 90.3, 66421 Homburg, Saar, Germany (J.B.-K.); (W.M.J.-W.)
| | - Joachim Oertel
- Department of Neurosurgery, Medical Faculty, Saarland University, Building 90.3, 66421 Homburg, Saar, Germany;
| | - Walter J. Schulz-Schaeffer
- Institute for Neuropathology, Medical Faculty, Saarland University, Building 90.3, 66421 Homburg, Saar, Germany (J.B.-K.); (W.M.J.-W.)
| |
Collapse
|
5
|
Fujiwara S. Clinical perspectives of rare ovarian tumors: clear cell ovarian cancer. Jpn J Clin Oncol 2023; 53:664-672. [PMID: 37288485 DOI: 10.1093/jjco/hyad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/16/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is a rare and distinct histological type of epithelial ovarian carcinoma in terms of its histopathological, clinical and genetic features. Patients with OCCC are younger and diagnosed at earlier stages than those with the most common histological type-high-grade serous carcinoma. Endometriosis is considered a direct precursor of OCCC. Based on preclinical data, the most frequent gene alternations in OCCC are mutations of AT-rich interaction domain 1A and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha. The prognosis of patients with early-stage OCCC is favorable, whereas patients at an advanced stage or who have the recurrent disease have a dismal prognosis due to OCCC's resistance to standard platinum-based chemotherapy. Despite a lower rate of response due to its resistance to standard platinum-based chemotherapy, the treatment strategy for OCCC resembles that of high-grade serous carcinoma, which includes aggressive cytoreductive surgery and adjuvant platinum-based chemotherapy. Alternative treatment strategies, including biological agents based on molecular characteristics specific to OCCC, are urgently needed. Furthermore, due to its rarity, well-designed collaborative international clinical trials are needed to improve oncologic outcomes and the quality of life in patients with OCCC.
Collapse
Affiliation(s)
- Satoe Fujiwara
- Department of Obstetrics and Gynecology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
6
|
Tong A, Di X, Zhao X, Liang X. Review the progression of ovarian clear cell carcinoma from the perspective of genomics and epigenomics. Front Genet 2023; 14:952379. [PMID: 36873929 PMCID: PMC9978161 DOI: 10.3389/fgene.2023.952379] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is a rare subtype of epithelial ovarian cancer with unique molecular characteristics, specific biological and clinical behavior, poor prognosis and high resistance to chemotherapy. Pushed by the development of genome-wide technologies, our knowledge about the molecular features of OCCC has been considerably advanced. Numerous studies are emerging as groundbreaking, and many of them are promising treatment strategies. In this article, we reviewed studies about the genomics and epigenetics of OCCC, including gene mutation, copy number variations, DNA methylation and histone modifications.
Collapse
Affiliation(s)
- An Tong
- Department of Gynecology and Obstetrics, Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangjie Di
- Clinical Trial Center, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Liang
- Department of Gynecology and Obstetrics, Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Mandal J, Mandal P, Wang TL, Shih IM. Treating ARID1A mutated cancers by harnessing synthetic lethality and DNA damage response. J Biomed Sci 2022; 29:71. [PMID: 36123603 PMCID: PMC9484255 DOI: 10.1186/s12929-022-00856-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
Chromatin remodeling is an essential cellular process for organizing chromatin structure into either open or close configuration at specific chromatin locations by orchestrating and modifying histone complexes. This task is responsible for fundamental cell physiology including transcription, DNA replication, methylation, and damage repair. Aberrations in this activity have emerged as epigenomic mechanisms in cancer development that increase tumor clonal fitness and adaptability amidst various selection pressures. Inactivating mutations in AT-rich interaction domain 1A (ARID1A), a gene encoding a large nuclear protein member belonging to the SWI/SNF chromatin remodeling complex, result in its loss of expression. ARID1A is the most commonly mutated chromatin remodeler gene, exhibiting the highest mutation frequency in endometrium-related uterine and ovarian carcinomas. As a tumor suppressor gene, ARID1A is essential for regulating cell cycle, facilitating DNA damage repair, and controlling expression of genes that are essential for maintaining cellular differentiation and homeostasis in non-transformed cells. Thus, ARID1A deficiency due to somatic mutations propels tumor progression and dissemination. The recent success of PARP inhibitors in treating homologous recombination DNA repair-deficient tumors has engendered keen interest in developing synthetic lethality-based therapeutic strategies for ARID1A-mutated neoplasms. In this review, we summarize recent advances in understanding the biology of ARID1A in cancer development, with special emphasis on its roles in DNA damage repair. We also discuss strategies to harness synthetic lethal mechanisms for future therapeutics against ARID1A-mutated cancers.
Collapse
Affiliation(s)
- Jayaprakash Mandal
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
8
|
Heinze K, Nazeran TM, Lee S, Krämer P, Cairns ES, Chiu DS, Leung SC, Kang EY, Meagher NS, Kennedy CJ, Boros J, Kommoss F, Vollert HW, Heitze F, du Bois A, Harter P, Grube M, Kraemer B, Staebler A, Kommoss FK, Heublein S, Sinn HP, Singh N, Laslavic A, Elishaev E, Olawaiye A, Moysich K, Modugno F, Sharma R, Brand AH, Harnett PR, DeFazio A, Fortner RT, Lubinski J, Lener M, Tołoczko-Grabarek A, Cybulski C, Gronwald H, Gronwald J, Coulson P, El-Bahrawy MA, Jones ME, Schoemaker MJ, Swerdlow AJ, Gorringe KL, Campbell I, Cook L, Gayther SA, Carney ME, Shvetsov YB, Hernandez BY, Wilkens LR, Goodman MT, Mateoiu C, Linder A, Sundfeldt K, Kelemen LE, Gentry-Maharaj A, Widschwendter M, Menon U, Bolton KL, Alsop J, Shah M, Jimenez-Linan M, Pharoah PD, Brenton JD, Cushing-Haugen KL, Harris HR, Doherty JA, Gilks B, Ghatage P, Huntsman DG, Nelson GS, Tinker AV, Lee CH, Goode EL, Nelson BH, Ramus SJ, Kommoss S, Talhouk A, Köbel M, Anglesio MS. Validated biomarker assays confirm that ARID1A loss is confounded with MMR deficiency, CD8 + TIL infiltration, and provides no independent prognostic value in endometriosis-associated ovarian carcinomas. J Pathol 2022; 256:388-401. [PMID: 34897700 PMCID: PMC9544180 DOI: 10.1002/path.5849] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/12/2021] [Accepted: 12/10/2021] [Indexed: 11/11/2022]
Abstract
ARID1A (BAF250a) is a component of the SWI/SNF chromatin modifying complex, plays an important tumour suppressor role, and is considered prognostic in several malignancies. However, in ovarian carcinomas there are contradictory reports on its relationship to outcome, immune response, and correlation with clinicopathological features. We assembled a series of 1623 endometriosis-associated ovarian carcinomas, including 1078 endometrioid (ENOC) and 545 clear cell (CCOC) ovarian carcinomas, through combining resources of the Ovarian Tumor Tissue Analysis (OTTA) Consortium, the Canadian Ovarian Unified Experimental Resource (COEUR), local, and collaborative networks. Validated immunohistochemical surrogate assays for ARID1A mutations were applied to all samples. We investigated associations between ARID1A loss/mutation, clinical features, outcome, CD8+ tumour-infiltrating lymphocytes (CD8+ TILs), and DNA mismatch repair deficiency (MMRd). ARID1A loss was observed in 42% of CCOCs and 25% of ENOCs. We found no associations between ARID1A loss and outcomes, stage, age, or CD8+ TIL status in CCOC. Similarly, we found no association with outcome or stage in endometrioid cases. In ENOC, ARID1A loss was more prevalent in younger patients (p = 0.012) and was associated with MMRd (p < 0.001) and the presence of CD8+ TILs (p = 0.008). Consistent with MMRd being causative of ARID1A mutations, in a subset of ENOCs we also observed an association with ARID1A loss-of-function mutation as a result of small indels (p = 0.035, versus single nucleotide variants). In ENOC, the association with ARID1A loss, CD8+ TILs, and age appears confounded by MMRd status. Although this observation does not explicitly rule out a role for ARID1A influence on CD8+ TIL infiltration in ENOC, given current knowledge regarding MMRd, it seems more likely that effects are dominated by the hypermutation phenotype. This large dataset with consistently applied biomarker assessment now provides a benchmark for the prevalence of ARID1A loss-of-function mutations in endometriosis-associated ovarian cancers and brings clarity to the prognostic significance. © 2021 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Karolin Heinze
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Tayyebeh M. Nazeran
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Sandra Lee
- University of Calgary, Department of Pathology and Laboratory Medicine, Calgary, AB, Canada
| | - Pauline Krämer
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University Hospital Tübingen, Department of Women’s Health, Tübingen, Germany
| | - Evan S. Cairns
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
| | - Derek S. Chiu
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Samuel C.Y. Leung
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Eun Young Kang
- University of Calgary, Department of Pathology and Laboratory Medicine, Calgary, AB, Canada
| | - Nicola S. Meagher
- University of New South Wales, Adult Cancer Program, Lowy Cancer Research Centre, Sydney, New South Wales, Australia
- University of New South Wales, School of Women’s and Children’s Health, Sydney, New South Wales, Australia
| | - Catherine J. Kennedy
- The University of Sydney, Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
- Westmead Hospital, Department of Gynaecological Oncology, Sydney, New South Wales, Australia
| | - Jessica Boros
- The University of Sydney, Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
- Westmead Hospital, Department of Gynaecological Oncology, Sydney, New South Wales, Australia
| | - Friedrich Kommoss
- Medizin Campus Bodensee, Institute of Pathology, Friedrichshafen, Germany
| | - Hans-Walter Vollert
- Medizin Campus Bodensee, Department of Gynecology and Obstetrics, Friedrichshafen, Germany
| | - Florian Heitze
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
| | - Andreas du Bois
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
| | - Philipp Harter
- Kliniken Essen Mitte, Department of Gynecology and Gynecologic Oncology, Essen, Germany
| | - Marcel Grube
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University Hospital Tübingen, Department of Women’s Health, Tübingen, Germany
| | - Bernhard Kraemer
- University Hospital Tübingen, Department of Women’s Health, Tübingen, Germany
| | - Annette Staebler
- University Hospital Tübingen, Institute of Pathology and Neuropathology, Tübingen, Germany
| | - Felix K.F. Kommoss
- University Hospital Heidelberg, Institute of Pathology, Heidelberg, Germany
| | - Sabine Heublein
- University Hospital Heidelberg and National Center for Tumor Diseases, Department of Obstetrics and Gynecology, Heidelberg, Germany
| | - Hans-Peter Sinn
- University Hospital Heidelberg, Institute of Pathology, Heidelberg, Germany
| | - Naveena Singh
- Barts Health National Health Service Trust, Department of Pathology, London, UK
| | - Angela Laslavic
- University of Pittsburgh School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, PA, USA
| | - Esther Elishaev
- University of Pittsburgh School of Medicine, Department of Pathology, PA, USA
| | - Alex Olawaiye
- University of Pittsburgh School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, PA, USA
| | - Kirsten Moysich
- Roswell Park Cancer Institute, Department of Cancer Prevention and Control, Buffalo, NY, USA
| | - Francesmary Modugno
- University of Pittsburgh School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, PA, USA
| | - Raghwa Sharma
- Westmead Hospital, Tissue Pathology and Diagnostic Oncology, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- Western Sydney University, Sydney, New South Wales, Australia
| | - Alison H. Brand
- Westmead Hospital, Department of Gynaecological Oncology, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Paul R. Harnett
- Westmead Hospital, Department of Gynaecological Oncology, Sydney, New South Wales, Australia
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, New South Wales, Australia
| | - Anna DeFazio
- The University of Sydney, Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
- Westmead Hospital, Department of Gynaecological Oncology, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, New South Wales, Australia
| | - Renée T. Fortner
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Jan Lubinski
- Pomeranian Medical University, Department of Genetics and Pathology, International Hereditary Cancer Centre, Szczecin, Poland
| | - Marcin Lener
- Pomeranian Medical University, Department of Genetics and Pathology, International Hereditary Cancer Centre, Szczecin, Poland
| | - Aleksandra Tołoczko-Grabarek
- Pomeranian Medical University, Department of Genetics and Pathology, International Hereditary Cancer Centre, Szczecin, Poland
| | - Cezary Cybulski
- Pomeranian Medical University, Department of Genetics and Pathology, International Hereditary Cancer Centre, Szczecin, Poland
| | - Helena Gronwald
- Pomeranian Medical University, Department of Propaedeutics, Physical Diagnostics and Dental Physiotherapy, Szczecin, Poland
| | - Jacek Gronwald
- Pomeranian Medical University, Department of Genetics and Pathology, International Hereditary Cancer Centre, Szczecin, Poland
| | - Penny Coulson
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | - Mona A El-Bahrawy
- Imperial College London, Department of Metabolism, Digestion and Reproduction, Hammersmith Hospital, London, UK
| | - Michael E. Jones
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | - Minouk J. Schoemaker
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
| | - Anthony J. Swerdlow
- The Institute of Cancer Research, Division of Genetics and Epidemiology, London, UK
- The Institute of Cancer Research, Division of Breast Cancer Research, London, UK
| | - Kylie L. Gorringe
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, Australia
- Peter MacCallum Cancer Centre, Women’s Cancer Program, Melbourne, Australia
| | - Ian Campbell
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, Australia
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, Research Division, Melbourne, Australia
| | - Linda Cook
- The University of New Mexico, Division of Epidemiology and Biostatistics, Albuquerque, NM, USA
| | - Simon A. Gayther
- Cedars-Sinai Medical Center, Center for Bioinformatics and Functional Genomics and the Cedars Sinai Genomics Core, Los Angeles, CA, USA
| | - Michael E. Carney
- John A. Burns School of Medicine, University of Hawaii, Honolulu, Department of Obstetrics and Gynecology, HI, USA
| | - Yurii B. Shvetsov
- University of Hawaii Cancer Center, Epidemiology Program, Honolulu, HI, USA
| | | | - Lynne R. Wilkens
- University of Hawaii Cancer Center, Epidemiology Program, Honolulu, HI, USA
| | - Marc T. Goodman
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Los Angeles, CA, USA
| | - Constantina Mateoiu
- Sahlgrenska Academy at Gothenburg University, Sahlgrenska Center for Cancer Research, Department of Obstetrics and Gynecology, Gothenburg, Sweden
| | - Anna Linder
- Sahlgrenska Academy at Gothenburg University, Sahlgrenska Center for Cancer Research, Department of Obstetrics and Gynecology, Gothenburg, Sweden
| | - Karin Sundfeldt
- Sahlgrenska Academy at Gothenburg University, Sahlgrenska Center for Cancer Research, Department of Obstetrics and Gynecology, Gothenburg, Sweden
| | - Linda E. Kelemen
- Medical University of South Carolina, Hollings Cancer Center and Department of Public Health Sciences, Charleston, SC, USA
| | - Aleksandra Gentry-Maharaj
- University College London, MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, London, UK
- University College London, Department of Women’s Cancer, Institute for Women’s Health, London, UK
| | | | - Usha Menon
- University College London, MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, London, UK
| | - Kelly L. Bolton
- Washington University School of Medicine, Department of Hematology and Oncology, Division of Oncology, St. Louis, MO, USA
| | - Jennifer Alsop
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Mitul Shah
- Addenbrookes Hospital, Department of Histopathology, Cambridge, UK
| | | | - Paul D.P. Pharoah
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - James D. Brenton
- University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Kara L. Cushing-Haugen
- Fred Hutchinson Cancer Research Center, Program in Epidemiology, Division of Public Health Sciences, Seattle, WA, USA
| | - Holly R. Harris
- Fred Hutchinson Cancer Research Center, Program in Epidemiology, Division of Public Health Sciences, Seattle, WA, USA
| | - Jennifer A. Doherty
- University of Utah, Huntsman Cancer Institute, Department of Population Health Sciences, Salt Lake City, UT, USA
| | - Blake Gilks
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Prafull Ghatage
- University of Calgary, Department of Oncology, Division of Gynecologic Oncology, Calgary, AB, Canada
| | - David G. Huntsman
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Gregg S. Nelson
- University of Calgary, Department of Oncology, Division of Gynecologic Oncology, Calgary, AB, Canada
| | - Anna V. Tinker
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
- University of British Columbia, Department of Medicine, Vancouver, BC, Canada
| | - Cheng-Han Lee
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, AB, Canada
| | - Ellen L. Goode
- Mayo Clinic, Department of Health Science Research, Division of Epidemiology, Rochester, MN, USA
| | - Brad H. Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC, Canada
| | - Susan J. Ramus
- University of New South Wales, Adult Cancer Program, Lowy Cancer Research Centre, Sydney, New South Wales, Australia
- University of New South Wales, School of Women’s and Children’s Health, Sydney, New South Wales, Australia
| | - Stefan Kommoss
- University Hospital Tübingen, Department of Women’s Health, Tübingen, Germany
| | - Aline Talhouk
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| | - Martin Köbel
- University of Calgary, Department of Pathology and Laboratory Medicine, Calgary, AB, Canada
| | - Michael S. Anglesio
- University of British Columbia, Department of Obstetrics and Gynecology, Vancouver, BC, Canada
- University of British Columbia, Vancouver General Hospital, and BC Cancer. British Columbia’s Gynecological Cancer Research Team (OVCARE), Vancouver, BC, Canada
| |
Collapse
|
9
|
Colarossi C, Picardo MC, Colarossi L, Deiana E, D'Agata C, Fichera C, Aiello E, Giannone G, Memeo L. Clear Cell Carcinoma Arising in an Abdominal Wall Cesarean Section Scar: A Case Report With Description of Pathological and Molecular Features. Front Surg 2021; 8:735381. [PMID: 34595205 PMCID: PMC8476878 DOI: 10.3389/fsurg.2021.735381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Clear cell carcinoma is a clinically and biologically distinct type of carcinoma predominantly encountered in the ovary and endometrium. In the ovary, it is frequently associated with endometriosis, which is a well-known risk factor. Endometriosis has often been described in the abdominal wall of women who had a cesarean section; however, malignant transformation is a very rare event, occurring in <1% of cases. The authors report a case involving a 52-year-old woman with an abdominal wall nodule at a cesarean section scar. Radiology revealed a mass, measuring 8 cm in size, in the abdominal wall suggestive of a soft tissue tumor. After resection, histology revealed the presence of clear, eosinophilic, and hobnail cells, which, together with immunohistochemical and molecular findings, enabled the diagnosis of clear cell carcinoma of the abdominal wall. The present report describes the clinical, radiological, pathological, and molecular features of an unusual abdominal lesion that represents a rare but challenging diagnosis.
Collapse
Affiliation(s)
- Cristina Colarossi
- Pathology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Maria Carolina Picardo
- Surgical Oncology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Lorenzo Colarossi
- Pathology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Enrica Deiana
- Surgical Oncology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Costanza D'Agata
- Surgical Oncology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Corrado Fichera
- Surgical Oncology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Eleonora Aiello
- Pathology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Giorgio Giannone
- Surgical Oncology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| | - Lorenzo Memeo
- Pathology Unit, Department of Experimental Oncology, Mediterranean Institute of Oncology, Catania, Italy
| |
Collapse
|
10
|
Evaluation of SWI/SNF Protein Expression by Immunohistochemistry in Ovarian Clear Cell Carcinoma. Int J Gynecol Pathol 2021; 40:156-164. [PMID: 32897960 DOI: 10.1097/pgp.0000000000000687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ovarian clear cell carcinomas (OCCC) are known to harbor ARID1A mutations, and several recent studies have described immunohistochemical loss of SMARCA2, SMARCA4, and SMARCB1 in a subset of tumors. We performed ARID1A, SMARCA2, SMARCA4, and SMARCB1 immunohistochemistry on 105 OCCCs to identify possible associations with clinicopathologic features and assess their prognostic value in these tumors. ARID1A, SMARCA4, and SMARCB1 were considered retained if any tumor cell nucleus stained while for SMARCA2, >5% of tumor nuclei were required to be positive. Patients had a mean age of 56 yr and tumors averaged 13 cm in size. Most patients (63%) had stage I tumors with 47% being alive and well, 41% dead from disease, 10% dead from other causes, and 3% alive with disease at last follow-up (mean 72 mo). Tumors showed an admixture of architectural patterns, but papillary was most frequent (49%). Stromal hyalinization was detected in 83% of OCCCs and a background precursor in 78%. High-grade atypia and/or oxyphilic cells were noted in 45% and 29% of tumors, respectively. All OCCCs expressed SMARCA4 and SMARCB1, but the absence of ARID1A was noted in 30% of tumors and SMARCA2 in 8%. ARID1A-retained OCCCs were associated with a dominant tubulocystic or solid pattern, but no other clinicopathologic features reached statistical significance. No switch/sucrose non-fermentable protein expression was predictive of prognosis. Additional studies with known mutational status of these proteins are warranted to better assess their prognostic utility and develop a standardized immunohistochemical scoring system.
Collapse
|
11
|
ARID1A mutation/ARID1A loss is associated with a high immunogenic profile in clear cell ovarian cancer. Gynecol Oncol 2021; 162:679-685. [PMID: 34272091 DOI: 10.1016/j.ygyno.2021.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/10/2021] [Accepted: 07/04/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVES ARID1A mutation is frequently found in clear cell ovarian cancer (CCC) and endometrioid ovarian cancer (EC). Anti-PD-1 monotherapy has been found to have limited efficacy in epithelial ovarian cancer; however, anti-PD-1 therapy showed significant clinical benefit in some CCC. We sought to define the relationship of ARID1A mutation/ARID1A expression to the immunogenic profile of different histologic subtypes of ovarian cancer. METHODS We performed next-generation sequencing of 160 cancer-related genes. Also, we analyzed the immunohistochemical status of ARID1A, PD-L1, and CD8 with survival in different histologic subtypes of ovarian cancer in a total of 103 cases. RESULTS ARID1A mutation was found in 0% of the high-grade serous ovarian cancer (HGSC) (n = 36), 41.5% of the CCC (n = 41), 45.0% of the EC (n = 20), and 33.3% of the mucinous ovarian cancer (MC) (n = 6) cases. ARID1A loss was found in 19.4% of the HGSC, 75.6% of the CCC, 60.0% of the EC and 0% of the MC cases. ARID1A mutation was found to be associated with high PD-L1 (p < 0.001) or CD8 levels (p < 0.001) in CCC but not in other histologic subtypes. Meanwhile, ARID1A loss was associated with high PD-L1 or CD8 levels in CCC (p < 0.001) and HGSC (p < 0.001) but not in EC and MC. In addition, ARID1A mutation was associated with high tumor mutation burden in CCC (p = 0.006). CONCLUSIONS ARID1A mutation/ARID1A expression is associated with immune microenvironmental factors in CCC but not in EC. ARID1A status can be a biomarker for selecting candidates for immune checkpoint blockade in CCC.
Collapse
|
12
|
SWI/SNF chromatin remodeling complex alterations in meningioma. J Cancer Res Clin Oncol 2021; 147:3431-3440. [PMID: 33715086 DOI: 10.1007/s00432-021-03586-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE While SWI/SNF chromatin remodeling complex alterations occur in approximately 20% of cancer, the frequency and potential impact on clinical outcomes in meningiomas remains to be comprehensively elucidated. METHODS A large series of 255 meningiomas from a single institution that was enriched for high grade and recurrent lesions was identified. We performed next-generation targeted sequencing of known meningioma driver genes, including NF2, AKT1, PIK3CA, PIK3R1, and SMO and SWI/SNF chromatin remodeling complex genes, including ARID1A, SMARCA4, and SMARCB1 in all samples. Clinical correlates focused on clinical presentation and patient outcomes are presented. RESULTS The series included 63 grade I meningiomas and 192 high-grade meningiomas, including 173 WHO grade II and 19 WHO grade III. Samples from recurrent surgeries comprised 37.3% of the series. A total of 41.6% meningiomas were from the skull base. NF2, AKT1, PIK3CA, PIK3R1, and SMO were mutated in 40.8, 7.1, 3.5, 3.9, and 2.4% of samples, respectively. ARID1A, SMARCA4, and SMARCB1 mutations were observed in 17.3, 3.5, and 5.1% of samples, respectively. A total of 68.2% of ARID1A-mutant meningiomas harbored a p.Gln1327del in-frame deletion. ARID1A mutations were seen in 19.1% of Grade I, 16.8% of Grade II, and 15.8% of Grade III meningiomas (P = 0.9, Fisher's exact). Median overall survival was 16.3 years (95% CI 10.9, 16.8). With multivariable analysis, the presence of an ARID1A mutation was significantly associated with a 7.421-fold increased hazard of death (P = 0.04). CONCLUSION ARID1A mutations occur with similar frequency between low and high-grade meningiomas, but ARID1A mutations are independently prognostic of worse prognosis beyond clinical and histopathologic features.
Collapse
|
13
|
Wang Y, Hoang L, Ji JX, Huntsman DG. SWI/SNF Complex Mutations in Gynecologic Cancers: Molecular Mechanisms and Models. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 15:467-492. [PMID: 31977292 DOI: 10.1146/annurev-pathmechdis-012418-012917] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The SWI/SNF (mating type SWItch/Sucrose NonFermentable) chromatin remodeling complexes interact with histones and transcription factors to modulate chromatin structure and control gene expression. These evolutionarily conserved multisubunit protein complexes are involved in regulating many biological functions, such as differentiation and cell proliferation. Genomic studies have revealed frequent mutations of genes encoding multiple subunits of the SWI/SNF complexes in a wide spectrum of cancer types, including gynecologic cancers. These SWI/SNF mutations occur at different stages of tumor development and are restricted to unique histologic types of gynecologic cancers. Thus, SWI/SNF mutations have to function in the appropriate tissue and cell context to promote gynecologic cancer initiation and progression. In this review, we summarize the current knowledge of SWI/SNF mutations in the development of gynecologic cancers to provide insights into both molecular pathogenesis and possible treatment implications for these diseases.
Collapse
Affiliation(s)
- Yemin Wang
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L3, Canada; , , .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada; .,Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia V6Z 2K8, Canada
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada;
| | - Jennifer X Ji
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L3, Canada; , , .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada;
| | - David G Huntsman
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L3, Canada; , , .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada; .,Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia V6Z 2K8, Canada
| |
Collapse
|
14
|
Yan T, Zhao B, Wu Q, Wang W, Shi J, Li D, Stovall DB, Sui G. Characterization of G-quadruplex formation in the ARID1A promoter. Int J Biol Macromol 2020; 147:750-761. [PMID: 31982538 DOI: 10.1016/j.ijbiomac.2020.01.210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/22/2022]
Abstract
As a member of the SWI/SNF family, ARID1A plays an essential role in modulating chromatin structure and gene expression. The tumor suppressive function of ARID1A has been well-defined and its downregulation in cancers is attributed to genomic deletion, DNA methylation and microRNA-mediated inhibition. In this study, we demonstrated that the negative strand of a C-rich region in the upstream vicinity of the human ARID1A transcription start site could form G-quadruplexes. Synthesized oligonucleotides based on the sequence of this region exhibited molar ellipticity at specific wavelengths characteristic of G-quadruplex structures in circular dichroism analyses. The formation of G-quadruplexes by these oligonucleotides were also proved by native polyacrylamide gel electrophoresis, DNA synthesis block assays, immunofluorescent staining and dimethyl sulfate footprinting studies. In reporter assays, mutations of the G-quadruplex forming sequence reduced ARID1A promoter-mediated transcription. Transfection of the oligonucleotide with the full length of G-quadruplex motif region, but not its partial sequences or the mutants, could both promote endogenous ARID1A expression and reduce cell proliferation.
Collapse
Affiliation(s)
- Ting Yan
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Bo Zhao
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Qiong Wu
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Wenmeng Wang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Jinming Shi
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Dangdang Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Daniel B Stovall
- College of Arts and Sciences, Winthrop University, Rock Hill, SC 29733, United States
| | - Guangchao Sui
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
15
|
Therapeutic preferability of gemcitabine for ARID1A-deficient ovarian clear cell carcinoma. Gynecol Oncol 2019; 155:489-498. [DOI: 10.1016/j.ygyno.2019.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
|
16
|
Hu HM, Zhao X, Kaushik S, Robillard L, Barthelet A, Lin KK, Shah KN, Simmons AD, Raponi M, Harding TC, Bandyopadhyay S. A Quantitative Chemotherapy Genetic Interaction Map Reveals Factors Associated with PARP Inhibitor Resistance. Cell Rep 2019; 23:918-929. [PMID: 29669295 PMCID: PMC5935461 DOI: 10.1016/j.celrep.2018.03.093] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/07/2018] [Accepted: 03/20/2018] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy is used to treat most cancer patients, yet our understanding of factors that dictate response and resistance to such drugs remains limited. We report the generation of a quantitative chemical-genetic interaction map in human mammary epithelial cells charting the impact of the knockdown of 625 genes related to cancer and DNA repair on sensitivity to 29 drugs, covering all classes of chemotherapy. This quantitative map is predictive of interactions maintained in other cell lines, identifies DNA-repair factors, predicts cancer cell line responses to therapy, and prioritizes synergistic drug combinations. We identify that ARID1A loss confers resistance to PARP inhibitors in cells and ovarian cancer patients and that loss of GPBP1 causes resistance to cisplatin and PARP inhibitors through the regulation of genes involved in homologous recombination. This map helps navigate patient genomic data and optimize chemotherapeutic regimens by delineating factors involved in the response to specific types of DNA damage.
Collapse
Affiliation(s)
- Hsien-Ming Hu
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xin Zhao
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA
| | - Swati Kaushik
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Antoine Barthelet
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin K Lin
- Clovis Oncology, Inc., Boulder, CO 80301, USA
| | - Khyati N Shah
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | - Sourav Bandyopadhyay
- Bioengineering and Therapeutic Sciences, Helen Diller Family Comprehensive Cancer Center and Institute for Computational Health Sciences. University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
17
|
Takenaka M, Köbel M, Garsed DW, Fereday S, Pandey A, Etemadmoghadam D, Hendley J, Kawabata A, Noguchi D, Yanaihara N, Takahashi H, Kiyokawa T, Ikegami M, Takano H, Isonishi S, Ochiai K, Traficante N, Gadipally S, Semple T, Vassiliadis D, Amarasinghe K, Li J, Mir Arnau G, Okamoto A, Friedlander M, Bowtell DDL. Survival Following Chemotherapy in Ovarian Clear Cell Carcinoma Is Not Associated with Pathological Misclassification of Tumor Histotype. Clin Cancer Res 2019; 25:3962-3973. [PMID: 30967419 DOI: 10.1158/1078-0432.ccr-18-3691] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/24/2019] [Accepted: 04/02/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Although ovarian clear cell carcinomas (OCCC) are commonly resistant to platinum-based chemotherapy, good clinical outcomes are observed in a subset of patients. The explanation for this is unknown but may be due to misclassification of high-grade serous ovarian cancer (HGSOC) as OCCC or mixed histology. EXPERIMENTAL DESIGN To discover potential biomarkers of survival benefit following platinum-based chemotherapy, we ascertained a cohort of 68 Japanese and Australian patients in whom progression-free survival (PFS) and overall survival (OS) could be assessed. We performed IHC reclassification of tumors, and targeted sequencing and immunohistochemistry of known driver genes. Exome sequencing was performed in 10 patients who had either unusually long survival (N = 5) or had a very short time to progression (N = 5). RESULTS The majority of mixed OCCC (N = 6, 85.7%) and a small proportion of pure OCCC (N = 3, 4.9%) were reclassified as likely HGSOC. However, the PFS and OS of patients with misclassified samples were similar to that of patients with pathologically validated OCCC. Absent HNF1B expression was significantly correlated with longer PFS and OS (P = 0.0194 and 0.0395, respectively). Mutations in ARID1A, PIK3CA, PPP2R1A, and TP53 were frequent, but did not explain length of PFS and OS. An exploratory exome analysis of patients with favorable and unfavorable outcomes did not identify novel outcome-associated driver mutations. CONCLUSIONS Survival benefit following chemotherapy in OCCC was not associated with pathological misclassification of tumor histotype. HNF1B loss may help identify the subset of patients with OCCC with a more favorable outcome.
Collapse
Affiliation(s)
- Masataka Takenaka
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, Foothill Medical Center, University of Calgary, Calgary, Canada
| | - Dale W Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dariush Etemadmoghadam
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Victoria, Australia
| | - Joy Hendley
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ayako Kawabata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Daito Noguchi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nozomu Yanaihara
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takako Kiyokawa
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirokuni Takano
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Seiji Isonishi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuhiko Ochiai
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | - Timothy Semple
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | | - Jason Li
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Michael Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Victoria, Australia
| | | |
Collapse
|
18
|
Caumanns JJ, Wisman GBA, Berns K, van der Zee AGJ, de Jong S. ARID1A mutant ovarian clear cell carcinoma: A clear target for synthetic lethal strategies. Biochim Biophys Acta Rev Cancer 2018; 1870:176-184. [PMID: 30025943 DOI: 10.1016/j.bbcan.2018.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/20/2022]
Abstract
SWI/SNF chromatin remodeling complexes play an important role in the epigenetic regulation of chromatin structure and gene transcription. Mutual exclusive subunits in the SWI/SNF complex include the DNA targeting members ARID1A and ARID1B as well as the ATPases SMARCA2 and SMARCA4. SWI/SNF complexes are mutated across many cancer types. The highest mutation incidence is found in ARID1A, primarily consisting of deleterious mutations. Current advances have reported synthetic lethal interactions with the loss of ARID1A in several cancer types. In this review, we discuss targets that are only important for tumor growth in an ARID1A mutant context. We focus on synthetic lethal strategies with ARID1A loss in ovarian clear cell carcinoma, a cancer with the highest ARID1A mutation incidence (46-57%). ARID1A directed lethal strategies that can be exploited clinically include targeting of the DNA repair proteins PARP and ATR, and the epigenetic factors EZH2, HDAC2, HDAC6 and BRD2.
Collapse
Affiliation(s)
- Joseph J Caumanns
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Katrien Berns
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Centre Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
19
|
Nasioudis D, Mastroyannis SA, Albright BB, Haggerty AF, Ko EM, Latif NA. Adjuvant chemotherapy for stage I ovarian clear cell carcinoma: Patterns of use and outcomes. Gynecol Oncol 2018; 150:14-18. [DOI: 10.1016/j.ygyno.2018.04.567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/23/2022]
|
20
|
Ji JX, Wang YK, Cochrane DR, Huntsman DG. Clear cell carcinomas of the ovary and kidney: clarity through genomics. J Pathol 2018; 244:550-564. [PMID: 29344971 DOI: 10.1002/path.5037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Clear cell ovarian carcinoma (CCOC) and clear cell renal cell carcinoma (ccRCC) both feature clear cytoplasm, owing to the accumulation of cytoplasmic glycogen. Genomic studies have demonstrated several mutational similarities between these two diseases, including frequent alterations in the chromatin remodelling SWI-SNF and cellular proliferation phosphoinositide 3-kinase-mammalian target of rapamycin pathways, as well as a shared hypoxia-like mRNA expression signature. Although many targeted treatment options have been approved for advanced-stage ccRCC, CCOC patients are still treated with conventional platinum and taxane chemotherapy, to which they are resistant. To determine the extent of similarity between these malignancies, we performed unsupervised clustering of mRNA expression data from these cancers. This review highlights the similarities and differences between these two clear cell carcinomas to facilitate knowledge translation within future research efforts. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jennifer X Ji
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yi Kan Wang
- Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| |
Collapse
|
21
|
The ARID1A, p53 and ß-Catenin statuses are strong prognosticators in clear cell and endometrioid carcinoma of the ovary and the endometrium. PLoS One 2018; 13:e0192881. [PMID: 29451900 PMCID: PMC5815611 DOI: 10.1371/journal.pone.0192881] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/31/2018] [Indexed: 12/18/2022] Open
Abstract
Aim The objective of this study was to evaluate the prognostic value of ARID1A, p53, p21, p16 and ß-Catenin in endometrioid and clear cell ovarian and endometrial carcinomas. Materials and methods 97 tumors were available for analysis of ARID1A, p53, p21, p16 and ß-Catenin with the techniques of tissue microarray and immunohistochemistry. 32 were ovarian carcinomas and 65 were endometrial carcinomas. Results Endometrioid ovarian carcinomas showed negative staining for ARID1A (a) and p21 (b), aberrant expression of p53 (c) and p16 (d) and ß-Catenin positive nuclear expression (e) respectively in 19% (a), 100% (b), 28.6% (c), 52.4% (d) and 4.8% (e) of all cases. In the group of clear cell ovarian carcinomas it was 63.6% (a), 100% (b), 81.8% (c), 54.5% (d) and 0% (e). For endometrioid uterine carcinomas it was 75.7% (a), 94.9% (b), 30.5% (c), 52.1% (d) and 6.8% (e) and for clear cell uterine carcinomas it was 8.6% (a), 100% (b), 50% (c), 100% (d) and 0% (e). Survival analysis showed that negative expression of ARID1A, p53 aberrant expression and ß-Catenin nuclear positive staining are independent negative prognosticators in both, clear cell and endometrioid carcinoma, regardless of ovarian or uterine origin. Cox-Regression analysis showed them again as negative prognostic factors. Furthermore, we found a significant correlation between ARID1A and ß-Catenin expression in endometrioid uterine tumors. Conclusion The analyzed gynaecological carcinoma showed a distinct expression scheme of proteins that are associated with tumor suppression. We may conclude that ARID1A, p53 and ß-Catenin are the strongest prognostic factors by analyzing a subgroup of tumor suppressor genes in clear cell and endometrioid subtypes of ovarian and endometrial cancer and may be used along with traditional morphological and clinical characteristics for prognosis.
Collapse
|
22
|
Okawa R, Banno K, Iida M, Yanokura M, Takeda T, Iijima M, Kunitomi-Irie H, Nakamura K, Adachi M, Umene K, Nogami Y, Masuda K, Kobayashi Y, Tominaga E, Aoki D. Aberrant chromatin remodeling in gynecological cancer. Oncol Lett 2017; 14:5107-5113. [PMID: 29113150 DOI: 10.3892/ol.2017.6891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/11/2017] [Indexed: 12/16/2022] Open
Abstract
Epigenetic regulatory mechanisms are a current focus in studies investigating cancer. Chromatin remodeling alters chromatin structure and regulates gene expression, and aberrant chromatin remodeling is involved in carcinogenesis. AT-rich interactive domain-containing protein 1A (ARID1A) and SWItch/sucrose non-fermentable-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 are remodeling factors that are mutated in numerous types of cancer. In gynecological cancer, ARID1A mutations have been identified in 46-57% of clear cell carcinoma and 30% of endometrioid carcinoma. Mutations of chromodomain helicase, DNA-binding protein 4 have been detected in 17-21% of endometrial serous cancer, and mutations of ARID1A and mixed-lineage leukemia 3 occur in 36 and 27% of uterine carcinosarcoma, respectively. These data suggest that aberrant chromatin remodeling is a potential cause of cancer, and have led to the development of novel proteins targeting these processes. Additional accumulation of information on the mechanisms of chromatin remodeling and markers for these events may promote personalized anticancer therapies.
Collapse
Affiliation(s)
- Ryuichiro Okawa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Miho Iida
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Megumi Yanokura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takashi Takeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Moito Iijima
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Haruko Kunitomi-Irie
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kanako Nakamura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masataka Adachi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kiyoko Umene
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuya Nogami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
23
|
Alldredge JK, Eskander RN. EZH2 inhibition in ARID1A mutated clear cell and endometrioid ovarian and endometrioid endometrial cancers. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2017; 4:17. [PMID: 29093822 PMCID: PMC5663065 DOI: 10.1186/s40661-017-0052-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/12/2017] [Indexed: 12/30/2022]
Abstract
Clear cell carcinoma and endometrioid adenocarcinoma are histologic subtypes of ovarian and uterine cancer that demonstrate unique clinical behavior but share common underlying genomic aberrations and oncogenic pathways. ARID1A mutations are more frequently identified in these tumors, in comparison to other gynecologic histologies, and loss of ARID1A tumor suppressor function is thought to be an essential component of carcinogenic transformation. Several therapeutic targets in ARID1A mutated cancers are in development, including EZH2 inhibitors. EZH2 facilitates epigenetic methylation to modulate gene expression, and both uterine and ovarian cancers show evidence of EZH2 over expression. EZH2 inhibition in ARID1A mutated tumors acts in a synthetically lethal manner to suppress cell growth and promote apoptosis, revealing a unique new therapeutic opportunity. Several phase 1 and 2 clinical trials of EZH2 inhibitors are ongoing currently and there is considerable promise in translational trials for utilization of this new targeted therapy, both to capitalize on ARID1A loss of function and to increase sensitivity to platinum-based adjuvant chemotherapies. This review will synthesize the molecular carcinogenesis of these malignancies and their unique clinical behavior, as a foundation for an emerging frontier of targeted therapeutics - the synergistic inhibition of EZH2 in ARID1A mutated cancers.
Collapse
Affiliation(s)
- Jill K. Alldredge
- University of California, 101 The City Drive South Orange, Irvine, CA 92868 USA
| | - Ramez N. Eskander
- University of California, San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92029-S0987 USA
| |
Collapse
|
24
|
Whole-genome sequencing revealed novel prognostic biomarkers and promising targets for therapy of ovarian clear cell carcinoma. Br J Cancer 2017; 117:717-724. [PMID: 28728166 PMCID: PMC5572180 DOI: 10.1038/bjc.2017.228] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022] Open
Abstract
Background: Ovarian clear cell carcinoma (OCCC) is mostly resistant to standard chemotherapy that results in poor patient survival. To understand the genetic background of these tumours, we performed whole-genome sequencing of OCCC tumours. Methods: Tumour tissue samples and matched blood samples were obtained from 55 Japanese women diagnosed with OCCC. Whole-genome sequencing was performed using the Illumina HiSeq platform according to standard protocols. Results: Alterations to the switch/sucrose non-fermentable (SWI/SNF) subunit, the phosphatidylinositol-3-kinase (PI3K)/Akt signalling pathway, and the receptor tyrosine kinase (RTK)/Ras signalling pathway were found in 51%, 42%, and 29% of OCCC tumours, respectively. The 3-year overall survival (OS) rate for patients with an activated PI3K/Akt signalling pathway was significantly higher than that for those with inactive pathway (91 vs 40%, hazard ratio 0.24 (95% confidence interval (CI) 0.10–0.56), P=0.0010). Similarly, the OS was significantly higher in patients with the activated RTK/Ras signalling pathway than in those with the inactive pathway (91 vs 53%, hazard ratio 0.35 (95% CI 0.13–0.94), P=0.0373). Multivariable analysis revealed that activation of the PI3K/Akt and RTK/Ras signalling pathways was an independent prognostic factor for patients with OCCC. Conclusions: The PI3K/Akt and RTK/Ras signalling pathways may be potential prognostic biomarkers for OCCC patients. Furthermore, our whole-genome sequencing data highlight important pathways for molecular and biological characterisations and potential therapeutic targeting in OCCC.
Collapse
|
25
|
Kawabata A, Yanaihara N, Nagata C, Saito M, Noguchi D, Takenaka M, Iida Y, Takano H, Yamada K, Iwamoto M, Kiyokawa T, Okamoto A. Prognostic impact of interleukin-6 expression in stage I ovarian clear cell carcinoma. Gynecol Oncol 2017; 146:609-614. [PMID: 28673661 DOI: 10.1016/j.ygyno.2017.06.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/12/2017] [Accepted: 06/20/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Ovarian clear cell carcinoma (OCCC) frequently presents at an early stage. In stage I OCCC, the prognosis differs according to substage. In particular, predictive biomarkers and new treatment strategies are needed for stage IC2/IC3 disease. We investigated tumor biology and prognostic factors for stage I OCCC from a clinicopathological perspective, including the expression of ARID1A and IL-6, which are considered critical for OCCC carcinogenesis. METHODS A retrospective cohort study of 192 patients with stage I OCCC treated at a single institution was performed. We calculated overall survival (OS) with respect to 12 clinicopathological parameters that included the unique and diverse histological features of OCCC. RESULTS The estimated 5-year OS rate in patients with all stage I OCCC was 88.9% during a median of 91months of follow-up. The multivariate analysis indicated that substage classification and IL-6 expression status were associated with poor OS (p=0.010 and p=0.027, respectively). Loss of ARID1A expression had no impact on survival; however, it was associated with substage (p=0.001), capsule rupture status (p=0.011), and ascites cytology (p=0.016). No clear association was found between ARID1A and IL-6 expressions. Histological findings, including the presence of endometriosis, adenofibroma, architectural pattern, and tumor cell type, showed no prognostic effects. CONCLUSIONS Both substage classification and IL-6 expression status may be independent prognostic factors in stage I OCCC. Therefore, IL-6 molecular stratification may be crucial in optimizing therapeutic strategies for early stage OCCC to improve survival.
Collapse
Affiliation(s)
- Ayako Kawabata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Nozomu Yanaihara
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Chie Nagata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan; Department of Education for Clinical Research, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Misato Saito
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Daito Noguchi
- Department of Obstetrics and Gynecology, The Jikei University Kashiwa Hospital, 163-1 Kashiwashita, Kashiwa-shi, Chiba 277-0004, Japan
| | - Masataka Takenaka
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yasushi Iida
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hirokuni Takano
- Department of Obstetrics and Gynecology, The Jikei University Kashiwa Hospital, 163-1 Kashiwashita, Kashiwa-shi, Chiba 277-0004, Japan
| | - Kyosuke Yamada
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masami Iwamoto
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Takako Kiyokawa
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
26
|
Liu G, Xu P, Fu Z, Hua X, Liu X, Li W, Zhang M, Wu J, Wen J, Xu J, Jia X. Prognostic and Clinicopathological Significance of ARID1A in Endometrium-Related Gynecological Cancers: A Meta-Analysis. J Cell Biochem 2017; 118:4517-4525. [PMID: 28466574 DOI: 10.1002/jcb.26109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
The tumor suppressor gene, AT Rich Interactive Domain 1A (ARID1A) mutation has been reported in a variety of cancers, especially the endometrium-related gynecological cancers, including the ovarian clear cell carcinoma, ovarian endometrioid carcinoma, and uterine endometrioid carcinoma. However, the prognostic value of ARID1A in endometrium-related gynecological cancers is still inconclusive. Therefore, we performed this meta-analysis to evaluate the clinical significance of ARID1A in endometrium-related gynecological cancers. By systematically searching all the relevant studies from Pubmed, Cochrane Library, and Web of Science up to September 2016, 11 studies with 1,432 patients were included. All the study characteristics and the prognostic data were extracted. Hazard ratios (HRs) and 95% confidence intervals (CIs) were pooled using the fixed-effect or random-effect model. Our results indicated that negative ARID1A expression predicted shorter Progression free survival (PFS, HR, 1.84; 95%CI, 1.32-2.57, P = 0.000) of patients with endometrium related gynecological cancers, especially the patiently with OCCC and the patients in Japan. Besides, a marginal trend towards the same direction was found in the Overall analysis (OS, HR, 1.34; 95%CI, 0.93-1.93, P = 0.112). Furthermore, the significant correlation was achieved between the negative ARID1A expression and the FIGO stage of endometrium-related gynecological cancers, but not the other characteristics. J. Cell. Biochem. 118: 4517-4525, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Guangquan Liu
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Pengfei Xu
- Nanjing Maternity and Child Health Medical Institute, Nanjing Maternity and Child, Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Ziyi Fu
- Nanjing Maternity and Child Health Medical Institute, Nanjing Maternity and Child, Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xiangdong Hua
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Xiaoguang Liu
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Wenqu Li
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Mi Zhang
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Jiacong Wu
- Nantong Maternity and Child Health Care Hospital, Nantong, 226081, China
| | - Juan Wen
- Nanjing Maternity and Child Health Medical Institute, Nanjing Maternity and Child, Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Juan Xu
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| | - Xuemei Jia
- Department of Obstetrics Gynecology, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical, University, Nanjing, 210004, China
| |
Collapse
|
27
|
Choi JY, Han HH, Kim YT, Lee JH, Kim BG, Kang S, Cho NH. Ovarian Clear Cell Carcinoma Sub-Typing by ARID1A Expression. Yonsei Med J 2017; 58:59-66. [PMID: 27873496 PMCID: PMC5122653 DOI: 10.3349/ymj.2017.58.1.59] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/03/2016] [Accepted: 06/08/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Loss of AT-rich DNA-interacting domain 1A (ARID1A) has been identified as a driving mutation of ovarian clear cell carcinoma (O-CCC), a triple-negative ovarian cancer that is intermediary between serous and endometrioid subtypes, in regards to molecular and clinical behaviors. However, about half of O-CCCs still express BAF250a, the protein encoded by ARID1A. Herein, we aimed to identify signatures of ARID1A-positive O-CCC in comparison with its ARID1A-negative counterpart. MATERIALS AND METHODS Seventy cases of O-CCC were included in this study. Histologic grades and patterns of primary tumor, molecular marker immunohistochemistry profiles, and clinical outcomes were analyzed. RESULTS Forty-eight (69%) O-CCCs did not express BAF250a, which were designated as "ARID1A-negative." The other 22 (31%) O-CCCs were designated as "ARID1A-positive." ARID1A-positive tumors were more likely to be histologically of high grades (41% vs. 10%, p=0.003), ERβ-positive (45% vs. 17%, p=0.011), and less likely to be HNF1β-positive (77% vs. 96%, p=0.016) and E-cadherin-positive (59% vs. 83%, p=0.028) than ARID1A-negative tumors. Patient age, parity, tumor stage were not significantly different in between the two groups. Cancer-specific survival was not significantly different either. CONCLUSION We classified O-CCCs according to ARID1A expression status. ARID1A-positive O-CCCs exhibited distinct immunohistochemical features from ARID1A-negative tumors, suggesting a different underlying molecular event during carcinogenesis.
Collapse
Affiliation(s)
- Jae Yoon Choi
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Ho Han
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Tae Kim
- Department of Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Hyun Lee
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Baek Gil Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Suki Kang
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
28
|
Lyu C, Zhang Y, Zhou X, Lang J. ARID1A gene silencing reduces the sensitivity of ovarian clear cell carcinoma to cisplatin. Exp Ther Med 2016; 12:4067-4071. [PMID: 28105136 DOI: 10.3892/etm.2016.3863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 10/05/2016] [Indexed: 12/19/2022] Open
Abstract
In ovarian clear cell carcinoma (OCCC), the mutation rate of the AT-rich interaction domain 1A (ARID1A) gene is 46-57%. However, the effects of ARID1A gene silencing by small interfering RNA (siRNA) on the sensitivity of OCCC to cisplatin have not been investigated. Thus, this study aimed to elucidate the association between ARID1A gene silencing and drug resistance in OCCC. Three pairs of ARID1A gene siRNA fragments (siRNA-1, siRNA-2 and siRNA-3) were designed and transiently transfected into ES2 OCCC cells using RNAi Max reagent. Western blotting results demonstrated that the transfection reduced ARID1A protein expression levels, with the siRNA-3 group having the lowest levels. The IC50 value, determined using a Cell Counting kit-8 assay, was significantly increased by siRNA-3 transfection compared with that in blank control and negative control groups. The cell survival rate following treatment with 50 µM cisplatin for 48 h was significantly increased in the siRNA-3 group compared with the blank control and negative control groups. Flow cytometric analysis revealed that the apoptosis rate for cisplatin-treated cells was significantly lower in cells with siRNA-3 transfection than in those without, and the apoptosis rate in siRNA-3-transfected cells was lower than that in the negative control group. Western blot analysis showed that the expression level of AKT in cisplatin-treated cells was significantly decreased compared with that in the negative control group, and the AKT expression level in cisplatin-treated cells was significantly higher with siRNA-3 transfection than without. Therefore, the results demonstrated that ARID1A siRNA efficiently decreased ARID1A expression, which reduced cisplatin chemosensitivity and cell apoptosis in ES2 OCCC cells via the regulation of AKT expression.
Collapse
Affiliation(s)
- Changshuai Lyu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yinglan Zhang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Xingnan Zhou
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
29
|
Kang HJ, Lee IS, Park YS, Ho WJ, Sohn D, Ahn JY, Yook JH, Kim BS. Biomarkers of EBV-positive Gastric Cancers: Loss of PTEN Expression is Associated with Poor Prognosis and Nodal Metastasis. Ann Surg Oncol 2016; 23:3684-3692. [DOI: 10.1245/s10434-016-5284-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Indexed: 12/13/2022]
|
30
|
Sato S, Itamochi H, Oumi N, Chiba Y, Oishi T, Shimada M, Sato S, Chikumi J, Nonaka M, Kudoh A, Komatsu H, Harada T, Sugiyama T. Establishment and characterization of a novel ovarian clear cell carcinoma cell line, TU-OC-2, with loss of ARID1A expression. Hum Cell 2016; 29:181-7. [PMID: 26960408 DOI: 10.1007/s13577-016-0138-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/24/2016] [Indexed: 01/23/2023]
Abstract
A new cell line of human ovarian clear cell carcinoma (CCC), TU-OC-2, was established and characterized. The cells were polygonal in shape, grew in monolayers without contact inhibition and were arranged in islands like pieces of a jigsaw puzzle. The chromosome numbers ranged from 41 to 96. A low rate of proliferation was observed and the doubling time was 37.5 h. The IC50 values of cisplatin, 7-ethyl-10-hydroxycamptothecin (SN38), which is an active metabolite of camptothecin, and paclitaxel were 7.7 μM, 17.7 nM and 301 nM, respectively. The drug sensitivity assay indicated that TU-OC-2 was sensitive to SN38, but resistant to cisplatin and paclitaxel. Mutational analysis revealed that TU-OC-2 cells have no mutations of PIK3CA in exons 9 and 20 and of TP53 in exons 4-9. We observed the loss of ARID1A protein expression in TU-OC-2 cells by western blot analysis and in the original tumor tissue by immunohistochemistry. This cell line may be useful for studying the chemoresistant mechanisms of CCC and exploring novel therapeutic targets such as the ARID1A-related signaling pathway.
Collapse
Affiliation(s)
- Seiya Sato
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka-City, Iwate, 020-8505, Japan
| | - Hiroaki Itamochi
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka-City, Iwate, 020-8505, Japan.
| | - Nao Oumi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Youhei Chiba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka-City, Iwate, 020-8505, Japan
| | - Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Shinya Sato
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Jun Chikumi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Michiko Nonaka
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Akiko Kudoh
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Hiroaki Komatsu
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Tasuku Harada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago-City, Tottori, 683-8504, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka-City, Iwate, 020-8505, Japan
| |
Collapse
|
31
|
Ye S, Yang J, You Y, Cao D, Huang H, Wu M, Chen J, Lang J, Shen K. Clinicopathologic Significance of HNF-1β, AIRD1A, and PIK3CA Expression in Ovarian Clear Cell Carcinoma: A Tissue Microarray Study of 130 Cases. Medicine (Baltimore) 2016; 95:e3003. [PMID: 26945423 PMCID: PMC4782907 DOI: 10.1097/md.0000000000003003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ovarian clear cell carcinoma (CCC) is a distinct histologic subtype with relatively poor survival. No prognostic or predictive molecular marker is currently available. Recent studies have shown that AT-rich interactive domain 1A (ARID1A) and phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) mutations are common genetic changes in ovarian CCC. Hepatocyte nuclear factor-1β (HNF-1β) expression has been proven to be highly sensitive and specific for clear cell histology. However, the correlations between these biomarkers and clinicopathologic variables and survival outcomes are controversial. The immunohistochemical analysis for HNF-1β, ARID1A, and PIK3CA was performed on a tissue microarray (TMA) consisting of 130 cases of ovarian CCC (237 tissue blocks) linked with clinical information. The immunostaining results were interpreted in a manner consistent with previous publications. The associations between biomarker expression and clinical and prognostic features were examined. All statistical analyses were conducted using 2-sided tests, and a value of P < 0.05 was considered significant. HNF-1β was expressed in 92.8% of all primary ovarian tumors, while the loss of ARID1A and PIK3CA was noted in 56.2% and 45.0%, respectively. Early-stage tumors tended to have high levels of HNF-1β immunoreactivity and expression of ARID1A (P = 0.02 and P = 0.03). Most patients (76.9%, 20/26) with concurrent endometriosis stained negative for ARID1A (P = 0.02). No relation was found between PIK3CA expression and clinical features. Low-level HNF-1β expression and loss of ARID1A were more commonly observed in patients with tumor recurrence (P = 0.02 and P < 0.001). Antibody expression was not associated with platinum-based chemotherapy response. Patients with negative ARID1A expression had worse survival outcome in terms of both overall survival (OS) and progression-free survival (PFS) (P = 0.03 and P = 0.01, respectively). On the contrary, patients with high-level HNF-1β were associated with good prognosis (P = 0.02 for OS and P = 0.01 for PFS). PIK3CA expression had no impact on survival. For univariate and multivariate analyses, only HNF-1β expression seemed to be a prognostic factor for favorable OS (P = 0.04). The loss of ARID1A was correlated with late-stage and endometriosis-associated tumors. The measurement of ARID1A expression might be a method to predict the risk of recurrence. Among the 3 biomarkers, only high-level HNF-1β expression proved to be a positive predictor for OS.
Collapse
Affiliation(s)
- Shuang Ye
- From the Department of Obstetrics and Gynecology (SY, JY, DC, HH, MW, JL, KS) and Department of Pathology (YY, JC), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Cho HD, Lee JE, Jung HY, Oh MH, Lee JH, Jang SH, Kim KJ, Han SW, Kim SY, Kim HJ, Bae SB, Lee HJ. Loss of Tumor Suppressor ARID1A Protein Expression Correlates with Poor Prognosis in Patients with Primary Breast Cancer. J Breast Cancer 2015; 18:339-46. [PMID: 26770240 PMCID: PMC4705085 DOI: 10.4048/jbc.2015.18.4.339] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/23/2015] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Somatic mutations of the chromatin remodeling AT-rich interactive domain 1A (SWI-like) gene (ARID1A) have been identified in many human cancers, including breast cancer. The purpose of this study was to evaluate the nuclear expression of ARID1A in breast cancers by immunohistochemistry (IHC) and to correlate the findings to clinicopathologic variables including prognostic significance. METHODS IHC was performed on tissue microarrays of 476 cases of breast cancer. Associations between ARID1A expression and clinicopathologic characteristics and molecular subtype were retrospectively analyzed. RESULTS Low expression of ARID1A was found in 339 of 476 (71.2%) cases. Low expression of ARID1A significantly correlated with positive lymph node metastasis (p=0.027), advanced pathologic stage (p=0.001), low Ki-67 labeling index (p=0.003), and negative p53 expression (p=0.017). The ARID1A low expression group had significantly shorter disease-free and overall survival than the ARID1A high expression group (p<0.001 and p<0.001, respectively). Multivariate analysis demonstrated that low expression of ARID1A was a significant independent predictive factor for poor disease-free and overall survival in patients with breast cancer (disease-free survival: hazard ratio, 0.38, 95% confidence interval [CI], 0.20-0.73, p=0.004; overall survival: hazard ratio, 0.11, 95% CI, 0.03-0.46, p=0.003). In patients with luminal A type disease, patients with low ARID1A expression had significantly shorter disease-free and overall survival rates than patients with high ARID1A expression (p=0.022 and p=0.018, respectively). CONCLUSION Low expression of ARID1A is an independent prognostic factor for disease-free and overall survival in breast cancer patients and may be associated with luminal A type disease. Although the biologic function of ARID1A in breast cancer remains unknown, low expression of ARID1A can provide valuable prognostic information.
Collapse
Affiliation(s)
- Hyun Deuk Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Jong Eun Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hae Yoen Jung
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Mee-Hye Oh
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Ji-Hye Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Si-Hyong Jang
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Kyung-Ju Kim
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sun Wook Han
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Yong Kim
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Han Jo Kim
- Department of Hemato-Oncology, Internal Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sang Byung Bae
- Department of Hemato-Oncology, Internal Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hyun Ju Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
33
|
Takeda T, Banno K, Okawa R, Yanokura M, Iijima M, Irie-Kunitomi H, Nakamura K, Iida M, Adachi M, Umene K, Nogami Y, Masuda K, Kobayashi Y, Tominaga E, Aoki D. ARID1A gene mutation in ovarian and endometrial cancers (Review). Oncol Rep 2015; 35:607-13. [PMID: 26572704 PMCID: PMC4689482 DOI: 10.3892/or.2015.4421] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/13/2015] [Indexed: 12/12/2022] Open
Abstract
The AT-rich interacting domain-containing protein 1A gene (ARID1A) encodes ARID1A, a member of the SWI/SNF chromatin remodeling complex. Mutation of ARID1A induces changes in expression of multiple genes (CDKN1A, SMAD3, MLH1 and PIK3IP1) via chromatin remodeling dysfunction, contributes to carcinogenesis, and has been shown to cause transformation of cells in association with the PI3K/AKT pathway. Information on ARID1A has emerged from comprehensive genome-wide analyses with next-generation sequencers. ARID1A mutations have been found in various types of cancer and occur at high frequency in endometriosis-associated ovarian cancer, including clear cell adenocarcinoma and endometrioid adenocarcinoma, and also occur at endometrial cancer especially in endometrioid adenocarcinoma. It has also been suggested that ARID1A mutation occurs at the early stage of canceration from endometriosis to endometriosis-associated carcinoma in ovarian cancer and also from atypical endo-metrial hyperplasia to endometrioid adenocarcinoma in endometrial cancer. Therefore, development of a screening method that can detect mutations of ARID1A and activation of the PI3K/AKT pathway might enable early diagnosis of endometriosis-associated ovarian cancers and endometrial cancers. Important results may also emerge from a current clinical trial examining a multidrug regimen of temsirolimus, a small molecule inhibitor of the PI3K/AKT pathway, for treatment of advanced ovarian clear cell adenocarcinoma with ARID1A mutation and PI3K/AKT pathway activation. Also administration of sorafenib, a multikinase inhibitor, can inhibit cancer proliferation with PIK3CA mutation and resistance to mTOR inhibitors and GSK126, a molecular-targeted drug can inhibit proliferation of ARID1A-mutated ovarian clear cell adenocarcinoma cells by targeting and inhibiting EZH2. Further studies are needed to determine the mechanism of chromatin remodeling dysregulation initiated by ARID1A mutation, to develop methods for early diagnosis, to investigate new cancer therapy targeting ARID1A, and to examine the involvement of ARID1A mutations in development, survival and progression of cancer cells.
Collapse
Affiliation(s)
- Takashi Takeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Ryuichiro Okawa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Megumi Yanokura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Moito Iijima
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Haruko Irie-Kunitomi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kanako Nakamura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Miho Iida
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Masataka Adachi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kiyoko Umene
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Yuya Nogami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| |
Collapse
|
34
|
Chromatin Remodelers: From Function to Dysfunction. Genes (Basel) 2015; 6:299-324. [PMID: 26075616 PMCID: PMC4488666 DOI: 10.3390/genes6020299] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/01/2015] [Accepted: 06/03/2015] [Indexed: 12/20/2022] Open
Abstract
Chromatin remodelers are key players in the regulation of chromatin accessibility and nucleosome positioning on the eukaryotic DNA, thereby essential for all DNA dependent biological processes. Thus, it is not surprising that upon of deregulation of those molecular machines healthy cells can turn into cancerous cells. Even though the remodeling enzymes are very abundant and a multitude of different enzymes and chromatin remodeling complexes exist in the cell, the particular remodeling complex with its specific nucleosome positioning features must be at the right place at the right time in order to ensure the proper regulation of the DNA dependent processes. To achieve this, chromatin remodeling complexes harbor protein domains that specifically read chromatin targeting signals, such as histone modifications, DNA sequence/structure, non-coding RNAs, histone variants or DNA bound interacting proteins. Recent studies reveal the interaction between non-coding RNAs and chromatin remodeling complexes showing importance of RNA in remodeling enzyme targeting, scaffolding and regulation. In this review, we summarize current understanding of chromatin remodeling enzyme targeting to chromatin and their role in cancer development.
Collapse
|