1
|
Prueksaritanond N, Petchsila K, Insin P. The Association of Preoperative Prognostic Nutritional Index With Survival Outcome in Ovarian Clear Cell Cancer. World J Oncol 2024; 15:950-959. [PMID: 39697428 PMCID: PMC11650612 DOI: 10.14740/wjon1963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/08/2024] [Indexed: 12/20/2024] Open
Abstract
Background The preoperative prognostic nutritional index (PPNI) has been investigated as a prognostic indicator in various cancers including epithelial ovarian cancer (EOC). However, its prognostic relevance in epithelial ovarian clear cell cancer (EOC-CC) remains uncertain. The objective of the study was to clarify the prognostic values of PPNI in EOC-CC patients. Methods We retrospectively reviewed 290 EOC-CC patients who underwent staging surgery at Rajavithi Hospital between January 2008 and December 2019. The PPNI was calculated using serum albumin × 10 (g/L) + 0.005 × peripheral blood lymphocyte count (per mm3). The association between PPNI and survival outcome was analyzed using the Kaplan-Meier method and the Cox proportional hazard model. Results The optimal cut-off value of PPNI, set at a mean of PPNI as 50, divided the EOC-CC patients into two groups: the low (n = 115) and the high (n = 175) PPNI group. With a median follow-up time of 63 months, patients with high PPNI exhibited significantly superior 5-year overall survival (OS) rates (76.4% vs. 56.8%, P = 0.004) and 5-year progression-free survival (PFS) rates (71.0% vs. 58.7%, P = 0.017) compared to patients with low PPNI. Univariate analysis revealed high PPNI correlated with increased OS (hazard ratio (HR): 0.51; 95% confidence interval (CI): 0.35 - 0.75) and PFS (HR: 0.63; 95% CI: 0.43 - 0.92). Nevertheless, in a multivariate analysis, high PPNI did not retain its status as an independent prognostic factor for a favorable prognosis in EOC-CC patients. Conclusion The present study did not confirm the prognostic significance of PPNI on survival outcomes in EOC-CC patients. Therefore, conducting prospective clinical research with large samples is necessary to illustrate the predictive values of PPNI in this rare disease.
Collapse
Affiliation(s)
- Nisa Prueksaritanond
- Gynecologic Oncology Unit, Department of Obstetrics and Gynecology, Rajavithi Hospital, Bangkok, Thailand
- College of Medicine, Rangsit University, Bangkok, Thailand
| | - Kittisak Petchsila
- Gynecologic Oncology Unit, Department of Obstetrics and Gynecology, Rajavithi Hospital, Bangkok, Thailand
| | - Putsarat Insin
- Gynecologic Oncology Unit, Department of Obstetrics and Gynecology, Rajavithi Hospital, Bangkok, Thailand
- College of Medicine, Rangsit University, Bangkok, Thailand
| |
Collapse
|
2
|
Xia S, Chen L, Yu M, Li J, Chen J, Xu F, Ni M, Liu C, Wu X, Chen X, Li J. Genetic and therapeutic heterogeneity shape the baseline and longitudinal immune ecosystem of ovarian clear cell carcinoma. J Immunother Cancer 2024; 12:e010069. [PMID: 39608974 PMCID: PMC11603735 DOI: 10.1136/jitc-2024-010069] [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: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) is a rare and chemo-resistant subtype of ovarian cancer. While immunotherapy has demonstrated effectiveness in some OCCC cases, the mechanisms for heterogeneous immunoreactivity and potential combinatory strategies remain unclear. METHODS Tumor samples from 13 patients with OCCC underwent single-cell mRNA-seq and TCR-seq to generate 1 40 683 cells transcriptome, while additionally 31 formalin-fixed paraffin-embedded samples were used for immunohistochemistry. Spatial transcriptomics of two OCCC samples and bulk RNA-seq of 58 patients were incorporated for spatial and interpatient level explorations. Serum tumor markers and radiologic images of three patients with OCCC who received combinatory VEGF and PD-1 inhibition were retrospectively analyzed. RESULTS OCCC exhibited a dynamic immune architecture shaped by genetic and therapeutic pressure. ARID1A mutation linked to baseline immune activation, correlated with an enrichment of neoantigen-reactive CXCL13+ CTLA4+ CD8+ T cells (p<0.001) and enhanced FASLG-FAS interactions. Recurrent OCCC was fibrotic, angiogenic, and immunosuppressive, exhibiting metabolic reprogramming towards activated activity in fatty acid metabolism. High CD36 (log-rank p=0.012, HR: 4.515) and CD47 expression (log-rank p=0.037, HR: 3.246) indicated worse progression-free survival. Treatment with bevacizumab increased intratumoral T cell infiltration and activated T cell interferon-γ signaling. Retrospective analysis of clinical cases revealed that combination therapy with anti-VEGF (vascular endothelial growth factor) and anti-PD-1 agents exerted clinical benefits in patients with OCCC with persistent, recurrent, and metastatic disease. CONCLUSIONS ARID1A mutation correlated with OCCC baseline immune activation. Stromal reconstruction and tumor metabolic reprogramming functioned as key processes of OCCC dynamic progression. VEGF inhibition remodeled OCCC stroma, restored T cell function and potentiated immunotherapy. CD36 and CD47 might be potential therapeutic targets for recurrent OCCC.
Collapse
Affiliation(s)
- Siyu Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Yu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiana Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiaxin Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Mengdong Ni
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chaohua Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
3
|
Yuan Z, Zhou H, Cao D, Yang J, Liu Q. Humanized patient-derived xenograft mouse model bearing ovarian clear cell carcinoma. J Gynecol Oncol 2024; 36:36.e40. [PMID: 39453393 DOI: 10.3802/jgo.2025.36.e40] [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: 03/06/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE The study aimed to establish humanized patient-derived xenograft (PDX) mouse models of ovarian clear cell carcinoma (OCCC) and evaluate their therapeutic responses. METHODS PDX models and their humanized counterparts (CD34+ humanized PDX models) derived from the same tumor source were developed, and the therapeutic responses were compared between the models. RESULTS Treatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor significantly reduced tumor size in traditional OCCC PDX models (p=0.021). Although differences in tumor growth between traditional PDX models and humanized PDX models were observed, they were not statistically significant (p=0.438). However, treatment effects of PI3K inhibitor differed significantly between conventional and humanized mice (p=0.006). In the Humanized PDX cohort, both programmed cell death protein-1 antibody monotherapy and PI3K inhibitor treatment slowed tumor growth relative to controls, with a synergistic effect noted during the latter part of the study, though these effects were not statistically significant. CONCLUSION This pioneering study successfully develop a humanized PDX model for OCCC, highlighting differential responses to treatments compared to conventional PDX models.
Collapse
Affiliation(s)
- Zhen Yuan
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Huimei Zhou
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Dongyan Cao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Southworth E, Thomson JP, Croy I, Churchman M, Arends MJ, Hollis RL, Gourley C, Herrington CS. Whole exome sequencing reveals diverse genomic relatedness between paired concurrent endometrial and ovarian carcinomas. Eur J Cancer 2024; 208:114205. [PMID: 38986422 DOI: 10.1016/j.ejca.2024.114205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Concurrent non-serous endometrial and ovarian tumours are often managed clinically as two separate primary tumours, but almost all exhibit evidence of a genomic relationship. METHODOLOGY This study investigates the extent of relatedness using whole exome sequencing, which was performed on paired non-serous endometrial and ovarian carcinomas from 27 patients with concurrent tumours in a cohort with detailed clinicopathological annotation. Four whole exome sequencing-derived parameters (mutation, mutational burden, mutational signatures and mutant allele tumour heterogeneity scores) were used to develop a novel unsupervised model for the assessment of genomic similarity to infer genomic relatedness of paired tumours. RESULTS This novel model demonstrated genomic relatedness across all four parameters in all tumour pairs. Mutations in PTEN, ARID1A, CTNNB1, KMT2D and PIK3CA occurred most frequently and 24 of 27 (89 %) tumour pairs shared identical mutations in at least one of these genes, with all pairs sharing mutations in a number of other genes. Ovarian endometriosis, CTNNB1 exon 3 mutation, and progression and death from disease were present across the similarity ranking. Mismatch repair deficiency was associated with less genomically similar pairs. DISCUSSION Although there was diversity across the cohort, the presence of genomic similarity in all paired tumours supports the hypothesis that concurrent non-serous endometrial and ovarian carcinomas are genomically related and may have arisen from a common origin.
Collapse
Affiliation(s)
- Emily Southworth
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - John P Thomson
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ian Croy
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Michael Churchman
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Robert L Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - C Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Pathology, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK.
| |
Collapse
|
6
|
Lopacinska-Jørgensen J, Vestergaard LK, Schejbel L, Høgdall CK, Poulsen TS, Høgdall EV. Paired comparison of the analytical performance between the Oncomine™ Comprehensive Assay v3 and whole-exome sequencing of ovarian cancer tissue. Mol Biol Rep 2024; 51:820. [PMID: 39017860 PMCID: PMC11255052 DOI: 10.1007/s11033-024-09715-y] [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: 02/19/2024] [Accepted: 06/09/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Next-generation sequencing (NGS) has been implemented in clinical oncology as a personalized medicine tool to identify targetable genetic alterations and to guide treatment decisions. However, the optimal NGS test strategy and target genes for clinical use are still being discussed. The aim was to compare the performance of the Oncomine™ Comprehensive Assay v3 (OCAv3) (targeted gene panel) and whole-exome sequencing (WES) to investigate somatic single and multiple nucleotide variants and small indels in ovarian cancer patients. METHODS AND RESULTS Genomic DNA was isolated from fresh frozen samples of five high-grade serous (HGSC) and three clear cell ovarian (oCCC) cancer patients. Exome sequencing libraries were prepared by using the Ion AmpliSeq Exome RDY kit, whereas libraries for OCAv3 were prepared using by Ion AmpliSeq™ Library Kit Plus. Sequencing was performed using the Ion S5XL System (Thermo Fisher Scientific). When including only variants classified as pathogenic, likely pathogenic or unknown significance based on ClinVar database verdicts and comparing overlapping regions covered both by the OCAv3 assay and WES, 23 variants were detected by both assays. However, OCAv3 detected additionally two variants: ARID1A: p.Gln563Ter and TP53: p.Ser261ValfsTer84 that have not passed WES filtering criteria due to low coverage. CONCLUSIONS With the present treatment possibilities, OCAv3 panel testing provided higher diagnostic yield due to better coverage. Our study emphasizes that WES, although offering the potential to identify novel findings in genes not covered by OCAv3, might overlook variants in genes relevant for OC.
Collapse
Affiliation(s)
- Joanna Lopacinska-Jørgensen
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 25, Herlev, 2730, Denmark
| | - Lau K Vestergaard
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 25, Herlev, 2730, Denmark
| | - Lone Schejbel
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 25, Herlev, 2730, Denmark
| | - Claus K Høgdall
- Department of Gynaecology, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tim Svenstrup Poulsen
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 25, Herlev, 2730, Denmark
| | - Estrid V Høgdall
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 25, Herlev, 2730, Denmark.
| |
Collapse
|
7
|
Hoshino H, Inoue D, Shinagawa A, Yoshida H, Shigeto S, Matsuda K, Akama TO, Yoshida Y, Kobayashi M. Establishment of a human ovarian clear cell carcinoma cell line mutant in PIK3CB but not PIK3CA. Hum Cell 2024; 37:1184-1193. [PMID: 38573494 PMCID: PMC11194194 DOI: 10.1007/s13577-024-01058-x] [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: 01/04/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
A human ovarian clear cell carcinoma cell line was established from a 46-year-old Japanese woman. That line, designated MTC-22, has proliferated continuously for over 6 months in conventional RPMI 1640 medium supplemented with 10% foetal bovine serum and has been passaged over 50 times. MTC-22 doubling-time is ~ 18 h, which is much shorter than most ovarian clear cell carcinoma lines reported to date. Morphologically, MTC-22 cells exhibit polygonal shapes and proliferate to form a monolayer in a jigsaw puzzle-like arrangement without contact inhibition. Ultrastructurally, cells exhibit numerous intracytoplasmic glycogen granules and well-developed mitochondria. G-band karyotype analysis indicated that cells have a complex karyotype close to tetraploid. We observed that the expression pattern of a series of ovarian carcinoma-related molecules in MTC-22 cells was identical to that seen in the patient's tumour tissue. Notably, MTC-22 cells, and the patient's carcinoma tissue, expressed low-sulphated keratan sulphate recognised by R-10G and 294-1B1 monoclonal antibodies, a hallmark of non-mucinous ovarian carcinoma, and particularly of clear cell ovarian carcinoma. Moreover, characteristic point mutations-one in ARID1A, which encodes the AT-rich interaction domain containing protein 1A, and the other in PIK3CB, which encodes the catalytic subunit of phosphoinositide 3-kinase-were seen in the patient's tumour tissue and retained in MTC-22 cells. Collectively, these findings indicate that MTC-22 cells could serve as a valuable tool for investigating the pathophysiology of ovarian clear cell carcinoma, particularly that harbouring PIK3CB mutations, and for developing and validating new diagnostic and therapeutic approaches to this life-threatening malignancy.
Collapse
Affiliation(s)
- Hitomi Hoshino
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Daisuke Inoue
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Akiko Shinagawa
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Hisato Yoshida
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Shohei Shigeto
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Kazuyuki Matsuda
- Department of Clinical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193, Japan.
| |
Collapse
|
8
|
Ueda M, Watanabe T, Momma T, Kanke Y, Kato A, Okabe C, Sato T, Kamo N, Endo Y, Furukawa S, Soeda S, Fujimori K. Diaphragmatic clear cell carcinoma with Lynch syndrome after surgery for atypical endometrial hyperplasia and ovarian endometriosis: A case report. Mol Clin Oncol 2024; 21:46. [PMID: 38826696 PMCID: PMC11140286 DOI: 10.3892/mco.2024.2744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/22/2024] [Indexed: 06/04/2024] Open
Abstract
Clear cell carcinoma (CCC) of the diaphragm is rare, with an origin that is reported to be associated with malignant transformation of extraperitoneal endometriosis. Lynch syndrome (LS) is an autosomal dominant hereditary cancer syndrome caused by germline pathogenic variants in one of the DNA mismatch repair (MMR) genes, MLH1, MSH2, MSH6 and PMS2. Women with LS have a significantly increased lifetime risk of endometrial and ovarian cancer. CCC is a common histology of endometriosis- and LS-associated malignancy. The present study describes the case of a 51-year-old woman with an intra-abdominal mass found during a routine physical examination. The patient had undergone total hysterectomy and bilateral adnexectomy for atypical endometrial hyperplasia (AEH) and ovarian endometriosis, respectively, 3 years previously. Enhanced computed tomography showed a mass on the surface of the liver. Laparoscopic examination of the abdominal cavity revealed a tumor on the underside of the right diaphragm, which was then surgically excised. Pathological examination of the excised tumor, along with immunohistochemistry, led to a diagnosis of CCC. Since LS was suspected due to the genetic family history of the patient, microsatellite instability analysis was performed on the diaphragmatic tumor, and the results were positive. Immunohistochemistry was performed for MMR proteins in AEH and CCC cells, both of which revealed loss of MSH2 and MSH6 expression. Following detailed genetic counseling, genetic testing of MMR genes was performed, revealing a germline pathogenic variant in MSH2 (c.1000C>T, p.Gln344*), thus confirming the diagnosis of LS. To the best of our knowledge, this is the first case report of concurrent diaphragmatic CCC and LS. Patients with LS and endometriosis are at risk of developing ovarian cancer or intra-abdominal malignant tumors. In addition, immunohistochemistry screening for MMR proteins should be considered in patients with AEH and a family history of LS-related cancer, to enable early clinical intervention in cases of endometrial cancer.
Collapse
Affiliation(s)
- Makiko Ueda
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Takafumi Watanabe
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Yasuyuki Kanke
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Asami Kato
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Chikako Okabe
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Tetsu Sato
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Norihito Kamo
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Yuta Endo
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Shigenori Furukawa
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Shu Soeda
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
9
|
Porter JM, McFarlane I, Bartos C, Churchman M, May J, Herrington CS, Connolly KC, Ryan NAJ, Hollis RL. The survival benefit associated with complete macroscopic resection in epithelial ovarian cancer is histotype specific. JNCI Cancer Spectr 2024; 8:pkae049. [PMID: 38902938 PMCID: PMC11233146 DOI: 10.1093/jncics/pkae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Complete macroscopic resection is a key factor associated with prolonged survival in ovarian cancer. However, most evidence derives from high-grade serous ovarian carcinoma, and the benefit of complete macroscopic resection in other histotypes is poorly characterized. We sought to determine which histotypes derive the greatest benefit from complete macroscopic resection to better inform future decisions on radical cytoreductive efforts. METHODS We performed multivariable analysis of disease-specific survival across 2 independent patient cohorts to determine the magnitude of benefit associated with complete macroscopic resection within each histotype. RESULTS Across both cohorts (Scottish: n = 1622; Surveillance, Epidemiology, and End Results [SEER]: n = 18 947), complete macroscopic resection was associated with prolonged disease-specific survival; this was more marked in the Scottish cohort (multivariable hazard ratio [HR] = 0.44, 95% confidence interval [CI] = 0.37 to 0.52 vs HR = 0.59, 95% CI = 0.57 to 0.62 in SEER). In both cohorts, clear cell ovarian carcinoma was among the histotypes to benefit most from complete macroscopic resection (multivariable HR = 0.23 and HR = 0.50 in Scottish and SEER cohorts, respectively); high-grade serous ovarian carcinoma patients demonstrated highly statistically significant and clinically meaningful survival benefit, but this was of lower magnitude than in clear cell ovarian carcinoma and endometrioid ovarian carcinoma across both cohorts. The benefit derived in low-grade serous ovarian carcinoma is also high (multivariable HR = 0.27 in Scottish cohort). Complete macroscopic resection was associated with prolonged survival in mucinous ovarian carcinoma patients in the SEER cohort (multivariable HR = 0.65), but the association failed to reach statistical significance in the Scottish cohort. CONCLUSIONS The overall ovarian cancer patient population demonstrates clinically significant survival benefit associated with complete macroscopic resection; however, the magnitude of benefit differs between histotypes.
Collapse
MESH Headings
- Humans
- Female
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/surgery
- Ovarian Neoplasms/pathology
- SEER Program
- Middle Aged
- Aged
- Carcinoma, Ovarian Epithelial/mortality
- Carcinoma, Ovarian Epithelial/surgery
- Carcinoma, Ovarian Epithelial/pathology
- Scotland/epidemiology
- Adenocarcinoma, Clear Cell/surgery
- Adenocarcinoma, Clear Cell/mortality
- Adenocarcinoma, Clear Cell/pathology
- Carcinoma, Endometrioid/mortality
- Carcinoma, Endometrioid/surgery
- Carcinoma, Endometrioid/pathology
- Cytoreduction Surgical Procedures/mortality
- Cystadenocarcinoma, Serous/surgery
- Cystadenocarcinoma, Serous/mortality
- Cystadenocarcinoma, Serous/pathology
- Adenocarcinoma, Mucinous/surgery
- Adenocarcinoma, Mucinous/mortality
- Adenocarcinoma, Mucinous/pathology
- Adult
- Neoplasms, Glandular and Epithelial/surgery
- Neoplasms, Glandular and Epithelial/mortality
- Neoplasms, Glandular and Epithelial/pathology
- Proportional Hazards Models
- Multivariate Analysis
- United States/epidemiology
Collapse
Affiliation(s)
- Joanna M Porter
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Iona McFarlane
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Clare Bartos
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Michael Churchman
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - James May
- The Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - C Simon Herrington
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- The Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - Kathryn C Connolly
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Neil A J Ryan
- The Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - Robert L Hollis
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
10
|
Hablase R, Kyrou I, Randeva H, Karteris E, Chatterjee J. The "Road" to Malignant Transformation from Endometriosis to Endometriosis-Associated Ovarian Cancers (EAOCs): An mTOR-Centred Review. Cancers (Basel) 2024; 16:2160. [PMID: 38893278 PMCID: PMC11172073 DOI: 10.3390/cancers16112160] [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: 04/19/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Ovarian cancer is an umbrella term covering a number of distinct subtypes. Endometrioid and clear-cell ovarian carcinoma are endometriosis-associated ovarian cancers (EAOCs) frequently arising from ectopic endometrium in the ovary. The mechanistic target of rapamycin (mTOR) is a crucial regulator of cellular homeostasis and is dysregulated in both endometriosis and endometriosis-associated ovarian cancer, potentially favouring carcinogenesis across a spectrum from benign disease with cancer-like characteristics, through an atypical phase, to frank malignancy. In this review, we focus on mTOR dysregulation in endometriosis and EAOCs, investigating cancer driver gene mutations and their potential interaction with the mTOR pathway. Additionally, we explore the complex pathogenesis of transformation, considering environmental, hormonal, and epigenetic factors. We then discuss postmenopausal endometriosis pathogenesis and propensity for malignant transformation. Finally, we summarize the current advancements in mTOR-targeted therapeutics for endometriosis and EAOCs.
Collapse
Affiliation(s)
- Radwa Hablase
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust Hospital, Guildford GU2 7XX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK (H.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 1GB, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Harpal Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK (H.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
| | - Jayanta Chatterjee
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust Hospital, Guildford GU2 7XX, UK
| |
Collapse
|
11
|
Collins K, Galea LA, Foroughi F, Siegmund SE, Anderson WJ, Appu S, Idrees MT, Ulbright TM, Acosta AM. Genomic analysis of primary epithelial neoplasms of the seminal vesicle identifies a subset of mucinous cystic tumours driven by KRAS mutations. Histopathology 2024; 84:1192-1198. [PMID: 38409850 DOI: 10.1111/his.15167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Carcinomas of the seminal vesicle are exceedingly rare, with a limited number of cases described in the literature. Reported cases span a relatively wide morphological spectrum, and their genomic features remain unexplored. DESIGN In this study, we interrogated five primary epithelial neoplasms of the seminal vesicle using a targeted DNA sequencing platform (OncoPanel, 447 genes). RESULTS The tumours included one adenocarcinoma with intestinal phenotype presenting after external beam radiation (for prostatic adenocarcinoma), one carcinoma with Müllerian-type clear cell phenotype, two mucinous tumours resembling low-grade mucinous neoplasms of the appendix (LAMN) and one mucinous cystadenoma. The post-radiation mucinous adenocarcinoma had genomic findings consistent with bi-allelic inactivation of TP53, as well as multiple copy-number changes with regional and chromosomal arm-level copy-number losses. The Müllerian-type clear cell carcinoma exhibited a complex copy-number profile with numerous regional and arm-level copy-number changes, as well as focal amplification events, including copy-number gain of 8q and amplification of a region within 20q13. Both low-grade mucinous tumours resembling LAMN harboured hot-spot gain-of-function KRAS variants (p.G12V and p.G13D) as the only genomic alteration. No genomic alterations were detected inthe lesion diagnosed as mucinous cystadenoma. CONCLUSION Our results suggest that primary low-grade mucinous neoplasms of the seminal vesicle may represent a distinct entity equivalent to appendiceal counterparts, driven by gain-of-function variants of RAS GTPases. The remaining tumours showed genomic features that closely resembled those of neoplasms with comparable phenotypes and/or biological characteristics arising in other sites, suggesting that they could be managed similarly, with special considerations related to their anatomical location.
Collapse
MESH Headings
- Humans
- Male
- Adult
- Aged
- Young Adult
- Middle Aged
- Proto-Oncogene Proteins p21(ras)/genetics
- Seminal Vesicles/pathology
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/pathology
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Cystadenoma, Mucinous/genetics
- Cystadenoma, Mucinous/pathology
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/pathology
Collapse
Affiliation(s)
- Katrina Collins
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laurence A Galea
- Department of Anatomical Pathology, Melbourne Pathology, Sonic Healthcare, Melbourne, VIC, Australia
| | - Forough Foroughi
- Department of Anatomical Pathology, QML Pathology, Brisbane, QLD, Australia
| | - Stephanie E Siegmund
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - William J Anderson
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sree Appu
- Department of Surgery, Monash University, Melbourne, VIC, Australia
| | - Muhammad T Idrees
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas M Ulbright
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andres M Acosta
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
12
|
Nomura S, Watanabe T, Honma R, Matsukura S, Ito E, Imai JI, Kiko Y, Suzuki O, Hashimoto Y, Kojima M, Furukawa S, Soeda S, Watanabe S, Fujimori K. Differentiation of ovarian serous carcinoma from ovarian clear cell carcinoma using a 10-gene signature selected by comprehensive gene expression analysis. Fukushima J Med Sci 2024; 70:65-73. [PMID: 38494731 PMCID: PMC11140200 DOI: 10.5387/fms.23-00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/17/2024] [Indexed: 03/19/2024] Open
Abstract
AIM Ovarian serous carcinoma (OSC) and ovarian clear cell carcinoma (OCCC) are two major histological types of epithelial ovarian carcinoma (EOC), each with different biological features and clinical behaviors. Although immunostaining is commonly used for differential diagnosis between OSC and OCCC, correct identification of EOC with mixed-type histology is sometimes a diagnostic challenge. The aim of the present study was to explore candidate genes as potential diagnostic biomarkers that distinguish OSC from OCCC. METHODS A total of 57 surgical specimens were obtained from EOC patients who had previously undergone primary debulking surgery. Total RNAs were extracted from fresh-frozen tissues of EOC patients, and were used for comprehensive gene expression analysis using DNA microarray technology. RESULTS Ten candidate genes, FXYD2, TMEM101, GABARAPL1, ARG2, GLRX, RBPMS, GDF15, PPP1R3B, TOB1, and GSTM3 were up-regulated in OCCC compared to OSC. All EOC patients were divided into two groups according to hierarchical clustering using a 10-gene signature. CONCLUSION Our data suggest that the 10 candidate genes would be an excellent marker for distinguishing OSC from OCCC. Furthermore, the molecular signatures of the 10 genes may enlighten us on the differences in carcinogenesis, and provide a theoretical basis for OCCC's resistance to chemotherapy in the future.
Collapse
Affiliation(s)
- Shinji Nomura
- Department of Obstetrics and Gynecology, Fukushima Medical University
| | - Takafumi Watanabe
- Department of Obstetrics and Gynecology, Fukushima Medical University
| | | | | | - Emi Ito
- Translational Research Center, Fukushima Medical University
| | - Jun-ichi Imai
- Translational Research Center, Fukushima Medical University
| | - Yuichiro Kiko
- Department of Diagnostic Pathology, Fukushima Medical University
| | - Osamu Suzuki
- Department of Diagnostic Pathology, Fukushima Medical University
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University
| | - Manabu Kojima
- Department of Obstetrics and Gynecology, Fukushima Medical University
| | | | - Shu Soeda
- Department of Obstetrics and Gynecology, Fukushima Medical University
| | | | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University
| |
Collapse
|
13
|
Qian J, Peng M, Li Y, Liu W, Zou X, Chen H, Zhou S, Xiao S, Zhou J. Case report: A germline CHEK1 c.613 + 2T>C leads to a splicing error in a family with multiple cancer patients. Front Oncol 2024; 14:1380093. [PMID: 38686193 PMCID: PMC11056527 DOI: 10.3389/fonc.2024.1380093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/21/2024] [Indexed: 05/02/2024] Open
Abstract
Background Genome instability plays a crucial role in promoting tumor development. Germline mutations in genes responsible for DNA repair are often associated with familial cancer syndromes. A noticeable exception is the CHEK1 gene. Despite its well-established role in homologous recombination, germline mutations in CHEK1 are rarely reported. Case presentation In this report, we present a patient diagnosed with ovarian clear cell carcinoma who has a family history of cancer. Her relatives include a grandfather with esophageal cancer, a father with gastric cancer, and an uncle with a brain tumor. The patient carried a typical genomic profile of clear cell carcinoma including mutations in KRAS, PPP2R1A, and PIK3R1. Importantly, her paired peripheral blood cells harbored a germline CHEK1 mutation, CHEK1 exon 6 c.613 + 2T>C, which was also found in her father. Unfortunately, the CHEK1 status of her grandfather and uncle remains unknown due to the unavailability of their specimens. Further evaluation via RT-PCR confirmed a splicing error in the CHEK1 gene, resulting in truncation at the kinase domain region, indicative of a loss-of-function mutation. Conclusion This case highlights a rare germline CHEK1 mutation within a family with a history of cancer. The confirmed splicing error at the mRNA level underscores the functional consequences of this mutation. Documenting such cases is vital for future evaluation of inheritance patterns, clinical penetrance of the mutation, and its association with specific cancer types.
Collapse
Affiliation(s)
- Jun Qian
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Peng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanan Li
- Molecular Genetics Laboratory, Suzhou Sano Precision Medicine Ltd., Suzhou, China
| | - Wei Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinwei Zou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huafei Chen
- Molecular Genetics Laboratory, Suzhou Sano Precision Medicine Ltd., Suzhou, China
| | - Sujuan Zhou
- Molecular Genetics Laboratory, Suzhou Sano Precision Medicine Ltd., Suzhou, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jinhua Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Liu Y, Zhao W, Huang C, Chu R, Li Z, Wang Y, Song L, Li L. The impact of lymphadenectomy on the survival outcomes of ovarian clear cell carcinoma: A retrospective study of the SEER database and Chinese registry. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:107975. [PMID: 38295548 DOI: 10.1016/j.ejso.2024.107975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/22/2023] [Accepted: 01/20/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) is a rare pathological type of ovarian cancer with a poor prognosis, and lymphadenectomy is controversial in patients with OCCC. The objective of this study was to evaluate the impact of lymphadenectomy on the prognosis of patients with OCCC. METHODS In this retrospective study, we collected data from the Surveillance, Epidemiology and End Results (SEER) database and institutional registries in China. The SEER cohort included 1777 women diagnosed with OCCC between 2010 and 2019, while the Chinese cohort included 199 women diagnosed between April 2004 and April 2021. Recurrence-free survival (RFS) and overall survival (OS) were studied using Kaplan-Meier curve and Cox regression analysis. We also employed propensity score matching (PSM) to adjust for baseline imbalances between the lymphadenectomy group and the no-lymphadenectomy group. RESULTS Multivariate cox regression analysis showed that lymphadenectomy was not associated with better overall survival (OS) in either early (hazard ratio [HR] 0.84[0.50-1.43], p = 0.528) or advanced (HR 0.78[0.50-1.21], p = 0.270) patients in the SEER cohort after PSM. Additionally, in the Kaplan-Meier curve analysis, lymphadenectomy did not significantly improve OS in both early (p = 0.28) and advanced (p = 0.49) patients in the SEER cohort after PSM. Similarly, in the Chinese cohort, lymphadenectomy had no significant effect on OS (early p = 0.22; advanced p = 0.61) or RFS (early p = 0.18; advanced p = 0.83) in both early and advanced patients. CONCLUSION In completely homogeneous groups, lymphadenectomy in women diagnosed with OCCC had no effect on either recurrence-free survival or overall survival compared to patients without lymphadenectomy.
Collapse
Affiliation(s)
- Yan Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Wenna Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Changzhen Huang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Ran Chu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China
| | - Zhuang Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Yuanjian Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Li Song
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Division of Gynecology Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China.
| | - Li Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
15
|
Ronchi S, Facchi S, Di Lauro E, Libera L, Carnevali IW, Zefiro F, Alexandrova E, Rizzo F, Sessa F, Tibiletti MG. Immunohistochemical and molecular pattern of p53 in epithelial ovarian cancers negative for germline BRCA1/2 variants. Pathol Res Pract 2024; 255:155183. [PMID: 38364651 DOI: 10.1016/j.prp.2024.155183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/18/2024]
Abstract
Epithelial ovarian cancers (EOC) associated with germline or somatic BRCA pathogenetic variants have a significantly higher rate of TP53aberrations. The majority of TP53 mutations are detectable by immunohistochemistry and several studies demonstrated that an abnormal p53 pattern characterized high-grade EOCs. An abnormal p53 immunohistochemical staining in fallopian tube (serous tubal intraepithelial carcinoma (STIC) and "p53 signature" is considered as a precancerous lesion of high-grade EOCs and it is often found in fallopian tube tissues of BRCA germline mutated patients suggesting that STIC is an early lesion and the TP53 mutation is an early driver event of BRCA mutated high-grade EOCs. No relevant data are present in literature about the involvement of p53 abnormal pattern in EOC carcinogenesis of patients negative for germline BRCA variants. We describe TP53 mutation results in relationship to the immunohistochemical pattern of p53 expression in a series of EOCs negative for BRCA1 and BRCA2 germline mutations. In addition, we also investigated STIC presence and "p53 signature" in fallopian tube sampling of these EOCs. Our results demonstrate that TP53 alterations are frequent and early events in sporadic EOCs including also low-grade carcinomas. Also in this series, STIC is associated with an abnormal p53 pattern in fallopian tubes of high-grade EOCs. In summary, TP53 aberrations are the most frequent and early molecular events in EOC carcinogenesis independently from BRCA mutation status.
Collapse
Affiliation(s)
- Susanna Ronchi
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Sofia Facchi
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Eleonora Di Lauro
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Laura Libera
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Ileana Wanda Carnevali
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Francesca Zefiro
- Department of Obstetric and Gynecology, ASST Settelaghi, University of Insubria, 21100 Varese, Italy.
| | - Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'ScuolaMedicaSalernitana', University of Salerno, 84081 Baronissi, Italy.
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'ScuolaMedicaSalernitana', University of Salerno, 84081 Baronissi, Italy; Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, 84081 Baronissi, SA, Italy.
| | - Fausto Sessa
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| | - Maria Grazia Tibiletti
- Unit of Pathology, Ospedale di Circolo, ASST SetteLaghi, Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy.
| |
Collapse
|
16
|
Akashi H, Yachida N, Ueda H, Yamaguchi M, Yamawaki K, Tamura R, Suda K, Ishiguro T, Adachi S, Nagase Y, Ueda Y, Ueda M, Abiko K, Kagabu M, Baba T, Nakaoka H, Enomoto T, Murai J, Yoshihara K. SLFN11 is a BRCA Independent Biomarker for the Response to Platinum-Based Chemotherapy in High-Grade Serous Ovarian Cancer and Clear Cell Ovarian Carcinoma. Mol Cancer Ther 2024; 23:106-116. [PMID: 37717249 DOI: 10.1158/1535-7163.mct-23-0257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/12/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
BRCA1/2 mutations are robust biomarkers for platinum-based chemotherapy in epithelial ovarian cancers. However, BRCA1/2 mutations in clear cell ovarian carcinoma (CCC) are less frequent compared with high-grade serous ovarian cancer (HGSC). The discovery of biomarkers that can be applied to CCC is an unmet need in chemotherapy. Schlafen 11 (SLFN11) has attracted attention as a novel sensitizer for DNA-damaging agents including platinum. In this study, we investigated the utility of SLFN11 in HGSC and CCC for platinum-based chemotherapy. SLFN11 expression was analyzed retrospectively by IHC across 326 ovarian cancer samples. The clinicopathologic significance of SLFN11 expression was analyzed across 57 advanced HGSC as a discovery set, 96 advanced HGSC as a validation set, and 57 advanced CCC cases, all of whom received platinum-based chemotherapy. BRCA1/2 mutation was analyzed using targeted-gene sequencing. In the HGSC cohort, the SLFN11-positive and BRCA mutation group showed significantly longer whereas the SLFN11-negative and BRCA wild-type group showed significantly shorter progression-free survival and overall survival. Moreover, SLFN11-positive HGSC shrunk significantly better than SLFN11-negative HGSC after neoadjuvant chemotherapy. Comparable results were obtained with CCC but without consideration of BRCA1/2 mutation due to a small population. Multivariate analysis identified SLFN11 as an independent factor for better survival in HGSC and CCC. The SLFN11-dependent sensitivity to platinum and PARP inhibitors were validated with genetically modified non-HGSC ovarian cancer cell lines. Our study reveals that SLFN11 predicts platinum sensitivity in HGSC and CCC independently of BRCA1/2 mutation status, indicating that SLFN11 assessment can guide treatment selection in HGSC and CCC.
Collapse
Affiliation(s)
- Hidehiko Akashi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nozomi Yachida
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Haruka Ueda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manako Yamaguchi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kaoru Yamawaki
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryo Tamura
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuaki Suda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sosuke Adachi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshikazu Nagase
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masashi Ueda
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation Chiyoda-ku, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
17
|
Marshall M, Elishaev E, Khader S. The diagnostic accuracy and prognostic implication of pelvic and peritoneal fluid cytology specimens in ovarian clear cell carcinoma. J Am Soc Cytopathol 2024; 13:53-58. [PMID: 37865568 DOI: 10.1016/j.jasc.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/23/2023]
Abstract
INTRODUCTION Ovarian clear cell carcinoma (OCCC) is a rare subtype of ovarian epithelial carcinoma. Patients with low-stage disease have an excellent prognosis, while the prognosis for those with high-stage disease is poor. Neoplastic cells in abdominopelvic washings upstages the patient to at least FIGO 1C3. Positive cytology confers a worse prognosis when compared to similar stage patients with negative cytology. This study aims to investigate the diagnostic performance of abdominopelvic fluid cytology specimens in cases with pure OCCC and reaffirm the importance of accurate cytologic detection and its impact on patient prognosis. MATERIALS AND METHODS The laboratory information system was queried to identify all patients treated for ovarian clear cell carcinoma at our institution over a period of 20 years with a companion abdominopelvic fluid cytology specimen at the time of surgical resection. Cases were sorted by the FIGO stage of the corresponding oophorectomy specimen. Cytology results, patient demographics, fluid volume, immunohistochemical results, and follow-up data were recorded. RESULTS A total of 143 cases were identified. The overall detection rate was 38%, with 54 of 143 cases positive for malignancy. Cytologic detection rates increased as FIGO stages increased. Fifty percent of stage 1C cases were upstaged on cytology alone. Ascites fluids performed better among stage 1 cases compared to pelvic wash specimens (77% detection rate versus 23%). Stage 1 patients with positive cytology trended towards a worse prognosis compared to those with negative cytology. CONCLUSIONS Positive cytology in low stage cases of OCCC has significant prognostic and therapeutic implications. Our large cohort further underscores the importance of accurate cytologic detection and subsequent staging in this setting.
Collapse
Affiliation(s)
- Mason Marshall
- Department of Pathology, Cytopathology Center of Excellence, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| | - Esther Elishaev
- Department of Pathology, Cytopathology Center of Excellence, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Samer Khader
- Department of Pathology, Cytopathology Center of Excellence, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
18
|
Hung SI, Chu MT, Hou MM, Lee YS, Yang CK, Chu SY, Liu FY, Hsu HC, Pao SC, Teng YC, Chen CB, Chao A, Chung WH, Chang JWC, Lai CH. Personalized neoantigen-based T cell therapy triggers cytotoxic lymphocytes expressing polyclonal TCR against metastatic ovarian cancer. Biomed Pharmacother 2023; 169:115928. [PMID: 38011788 DOI: 10.1016/j.biopha.2023.115928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Neoantigen-reactive cytotoxic T lymphocytes play a vital role in precise cancer cell elimination. In this study, we demonstrate the effectiveness of personalized neoantigen-based T cell therapy in inducing tumor regression in two patients suffering from heavily-burdened metastatic ovarian cancer. Our approach involved the development of a robust pipeline for ex vivo expansion of neoantigen-reactive T lymphocytes. Neoantigen peptides were designed and synthesized based on the somatic mutations of the tumors and their predicted HLA binding affinities. These peptides were then presented to T lymphocytes through co-culture with neoantigen-loaded dendritic cells for ex vivo expansion. Subsequent to cell therapy, both patients exhibited significant reductions in tumor marker levels and experienced substantial tumor regression. One patient achieved repeated cancer regression through infusions of T cell products generated from newly identified neoantigens. Transcriptomic analyses revealed a remarkable increase in neoantigen-reactive cytotoxic lymphocytes in the peripheral blood of the patients following cell therapy. These cytotoxic T lymphocytes expressed polyclonal T cell receptors (TCR) against neoantigens, along with abundant cytotoxic proteins and pro-inflammatory cytokines. The efficacy of neoantigen targeting was significantly associated with the immunogenicity and TCR polyclonality. Notably, the neoantigen-specific TCR clonotypes persisted in the peripheral blood after cell therapy. Our findings indicate that personalized neoantigen-based T cell therapy triggers cytotoxic lymphocytes expressing polyclonal TCR against ovarian cancer, suggesting its promising potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Shuen-Iu Hung
- Cancer Vaccine & Immune Cell Therapy Core Lab, Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Section 2, Linong Street, Taipei 112, Taiwan.
| | - Mu-Tzu Chu
- Cancer Vaccine & Immune Cell Therapy Core Lab, Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan.
| | - Ming-Mo Hou
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan.
| | - Yun-Shien Lee
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Guishan, Taoyuan 333, Taiwan; Department of Biotechnology, Ming-Chuan University, Taoyuan 333, Taiwan.
| | - Chan-Keng Yang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan.
| | - Sung-Yu Chu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan.
| | - Feng-Yuan Liu
- College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan; Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; School of Medicine, National Tsing Hua University, No.101, Section 2, Kuang-Fu Road, Hsinchu 300, Taiwan.
| | - Hung-Chih Hsu
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan.
| | - Shih-Cheng Pao
- Cancer Vaccine & Immune Cell Therapy Core Lab, Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, No.155, Section 2, Linong Street, Taipei 112, Taiwan.
| | - Yu-Chuan Teng
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Guishan, Taoyuan 333, Taiwan.
| | - Chun-Bing Chen
- College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan; Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan.
| | - Angel Chao
- College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan.
| | - Wen-Hung Chung
- Cancer Vaccine & Immune Cell Therapy Core Lab, Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan; Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung Branch, No. 222, Maijin Road., Keelung 204, Taiwan.
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan.
| | - Chyong-Huey Lai
- College of Medicine, Chang Gung University, No. 5, De-Ming Road., Taoyuan 333, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, No. 5. Fuxing Street, Taoyuan 333, Taiwan.
| |
Collapse
|
19
|
Li JJ, Lee CS. The Role of the AT-Rich Interaction Domain 1A Gene ( ARID1A) in Human Carcinogenesis. Genes (Basel) 2023; 15:5. [PMID: 38275587 PMCID: PMC10815128 DOI: 10.3390/genes15010005] [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: 11/13/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The switch/sucrose non-fermentable (SWI/SNF) (SWI/SNF) complex uses energy from ATP hydrolysis to mobilise nucleosomes on chromatin. Components of SWI/SNF are mutated in 20% of all human cancers, of which mutations in AT-rich binding domain protein 1A (ARID1A) are the most common. ARID1A is mutated in nearly half of ovarian clear cell carcinoma and around one-third of endometrial and ovarian carcinomas of the endometrioid type. This review will examine in detail the molecular functions of ARID1A, including its role in cell cycle control, enhancer regulation, and the prevention of telomerase activity. ARID1A has key roles in the maintenance of genomic integrity, including DNA double-stranded break repair, DNA decatenation, integrity of the cohesin complex, and reduction in replication stress, and is also involved in mismatch repair. The role of ARID1A loss in the pathogenesis of some of the most common human cancers is discussed, with a particular emphasis on gynaecological cancers. Finally, several promising synthetic lethal strategies, which exploit the specific vulnerabilities of ARID1A-deficient cancer cells, are briefly mentioned.
Collapse
Affiliation(s)
- Jing Jing Li
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
| | - Cheok Soon Lee
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2010, Australia
| |
Collapse
|
20
|
Yoshikawa N, Matsukawa T, Hattori S, Iyoshi S, Yoshida K, Yoshihara M, Tamauchi S, Shimizu Y, Ikeda Y, Yokoi A, Niimi K, Kawai M, Kajiyama H. Mean platelet volume as a potential biomarker for survival outcomes in ovarian clear cell carcinoma. Int J Clin Oncol 2023; 28:1680-1689. [PMID: 37804356 DOI: 10.1007/s10147-023-02417-8] [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: 06/13/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVE This study aimed to explore the prognostic value of mean platelet volume (MPV) in patients with ovarian clear cell carcinoma (OCCC) and evaluate the predictive performance of a random forest model incorporating MPV and other key clinicopathological factors. METHODS A total of 204 patients with OCCC treated between January 2004 and December 2019 were retrospectively analyzed. Clinicopathological characteristics and preoperative laboratory data were collected, and survival outcomes were evaluated using the Kaplan-Meier method and Cox proportional hazards models. An optimal MPV cutoff was determined by receiver operating characteristic (ROC) curve analysis. A random forest model was then constructed using the identified independent prognostic factors, and its predictive performance was evaluated. RESULTS The ROC analysis identified 9.3 fL as the MPV cutoff value for predicting 2-year survival. The MPV-low group had lower 5-year overall survival and progression-free survival rates than the MPV-high group (p = 0.003 and p = 0.034, respectively). High MPV emerged as an independent prognostic factor (p = 0.006). The random forest model, incorporating the FIGO stage, residual tumors, peritoneal cytology, and MPV, demonstrated robust predictive performance (area under the curve: 0.905). CONCLUSION MPV is a promising prognostic indicator in OCCC. Lower MPV correlated with worse survival rates, advocating its potential utility in refining patient management strategies. The commendable predictive performance of the random forest model, integrating MPV and other significant prognostic factors, suggests a pathway toward enhanced survival prediction, thereby warranting further research.
Collapse
Affiliation(s)
- Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Tetsuya Matsukawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Satomi Hattori
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yusuke Shimizu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kaoru Niimi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Michiyasu Kawai
- Department of Obstetrics and Gynecology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
21
|
Veneziani AC, Gonzalez-Ochoa E, Alqaisi H, Madariaga A, Bhat G, Rouzbahman M, Sneha S, Oza AM. Heterogeneity and treatment landscape of ovarian carcinoma. Nat Rev Clin Oncol 2023; 20:820-842. [PMID: 37783747 DOI: 10.1038/s41571-023-00819-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/04/2023]
Abstract
Ovarian carcinoma is characterized by heterogeneity at the molecular, cellular and anatomical levels, both spatially and temporally. This heterogeneity affects response to surgery and/or systemic therapy, and also facilitates inherent and acquired drug resistance. As a consequence, this tumour type is often aggressive and frequently lethal. Ovarian carcinoma is not a single disease entity and comprises various subtypes, each with distinct complex molecular landscapes that change during progression and therapy. The interactions of cancer and stromal cells within the tumour microenvironment further affects disease evolution and response to therapy. In past decades, researchers have characterized the cellular, molecular, microenvironmental and immunological heterogeneity of ovarian carcinoma. Traditional treatment approaches have considered ovarian carcinoma as a single entity. This landscape is slowly changing with the increasing appreciation of heterogeneity and the recognition that delivering ineffective therapies can delay the development of effective personalized approaches as well as potentially change the molecular and cellular characteristics of the tumour, which might lead to additional resistance to subsequent therapy. In this Review we discuss the heterogeneity of ovarian carcinoma, outline the current treatment landscape for this malignancy and highlight potentially effective therapeutic strategies in development.
Collapse
Affiliation(s)
- Ana C Veneziani
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Eduardo Gonzalez-Ochoa
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Husam Alqaisi
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ainhoa Madariaga
- Medical Oncology Department, 12 De Octubre University Hospital, Madrid, Spain
| | - Gita Bhat
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Marjan Rouzbahman
- Department of Laboratory Medicine and Pathobiology, Toronto General Hospital, Toronto, Ontario, Canada
| | - Suku Sneha
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Amit M Oza
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Tsuchimochi S, Wada-Hiraike O, Urano Y, Kukita A, Yamaguchi K, Honjo H, Taguchi A, Tanikawa M, Sone K, Mori-Uchino M, Tsuruga T, Oda K, Osuga Y. Characterization of a fluorescence imaging probe that exploits metabolic dependency of ovarian clear cell carcinoma. Sci Rep 2023; 13:20292. [PMID: 37985723 PMCID: PMC10662153 DOI: 10.1038/s41598-023-47637-0] [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: 07/10/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The purpose of this study is to clarify the metabolic dependence of ovarian clear cell carcinoma (CCC) by comparing normal tissues and to examine the applicability of fluorescence imaging probe to exploit these metabolic differences. Enhanced glutathione synthesis was supported by the increased uptake of related metabolites and elevated expression levels of genes. Accumulation of intracellular iron and lipid peroxide, induction of cell death by inhibition of the glutathione synthesis pathway indicated that ferroptosis was induced. The activation of γ-glutamyl hydroxymethyl rhodamine green (gGlu-HMRG), a fluorescent imaging probe that recognizes γ-glutamyl transferase, which is essential for the synthesis of glutathione, was investigated in fresh-frozen surgical specimens. gGlu-HMRG detected extremely strong fluorescent signals in the tumor lesions of CCC patients, compared to normal ovaries or endometrium. These results revealed that CCC occurs in the stressful and unique environment of free radical-rich endometrioma, and that glutathione metabolism is enhanced as an adaptation to oxidative stress. Furthermore, a modality that exploits these metabolic differences would be useful for distinguishing between CCC and normal tissues.
Collapse
Affiliation(s)
- Saki Tsuchimochi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan.
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
- CREST, Japan Agency for Medical Research and Development, Chiyoda, Tokyo, 100-0004, Japan
| | - Asako Kukita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Kohei Yamaguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Harunori Honjo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Mayuyo Mori-Uchino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Tetsushi Tsuruga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Katsutoshi Oda
- Department of Integrated Genomics, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, 113-8655, Japan
| |
Collapse
|
23
|
Kim YN, Chung YS, Lee JH, Park E, Lee ST, Kim S, Lee JY. Application of precision medicine based on next-generation sequencing and immunohistochemistry in ovarian cancer: a real-world experience. J Gynecol Oncol 2023; 34:e70. [PMID: 37417298 PMCID: PMC10627761 DOI: 10.3802/jgo.2023.34.e70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/02/2023] [Accepted: 05/13/2023] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVE To evaluate the landscape of gene alterations and immunohistochemistry (IHC) profiles of patients with ovarian cancer for targeted therapy and investigate the real-world experience of applying precision medicine. METHODS Patients diagnosed with ovarian cancer between January 2015 and May 2021 at Severance Hospital and who underwent tumor next-generation sequencing (NGS) were reviewed. Data on germline mutation, IHC markers for mismatch repair deficiency (MMRd), programmed death ligand 1 (PD-L1) expression, and human epidermal growth factor receptor 2 (HER2) expression were acquired. The use of matched therapy and its clinical outcomes were evaluated. RESULTS Of the 512 patients who underwent tumor NGS, 403 underwent panel-based germline testing. In patients who underwent both tests, tumor NGS identified 39 patients (9.7%) with BRCA mutations and 16 patients (4.0%) with other homologous recombination repair (HRR)-associated gene mutations, which were not found in germline testing. The most common single nucleotide variants were TP53 (82.2%), ARID1A (10.4%), PIK3CA (9.7%), and KRAS (8.4%). Copy number aberrations were found in 122 patients. MMRd was found in 3.2% of patients, high PD-L1 expression in 10.1%, and HER2 overexpression in 6.5%. Subsequently, 75 patients (14.6%) received a poly (ADP-ribose) polymerase inhibitor based on BRCA mutation and 11 patients (2.1%) based on other HRR-associated gene mutations. Six patients (1.2%) with MMRd underwent immunotherapy. Twenty-eight patients (5.5%) received other matched therapies targeting HER2, fibroblast growth factor receptor, folate receptor alpha, RAS, and PIK3CA. CONCLUSION A comprehensive review of germline mutation, IHC, and tumor NGS helped identify candidates for precision therapy in patients with ovarian cancer, a proportion of whom received matched therapy.
Collapse
Affiliation(s)
- Yoo-Na Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yun Soo Chung
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Eunhyang Park
- Department of Pathology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
24
|
Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2023; 49:2563-2574. [PMID: 37565583 DOI: 10.1111/jog.15761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
AIM This review aimed to describe the potential for therapeutic targeting of the JAK/STAT signaling pathway by repurposing the clinically-approved JAK inhibitor ruxolitinib in the patients with epithelial ovarian cancer (OC) setting. METHODS We reviewed publications that focus on the inhibition of the JAK/STAT pathway in hematological and solid malignancies including OC. RESULTS Preclinical studies showed that ruxolitinib effectively reduces OC cell viability and metastasis and enhances the anti-tumor activity of chemotherapy drugs. There are a number of recent clinical trials exploring the role of JAK/STAT inhibition in solid cancers including OC. Early results have not adequately supported efficacy in solid tumors. However, there are preclinical data and clinical studies supporting the use of ruxolitinib in combination with both chemotherapy and other targeted drugs in OC setting. CONCLUSION Inflammatory conditions and persistent activation of the JAK/STAT pathway are associated with tumourigenesis and chemoresistance, and therapeutic blockade of this pathway shows promising results. For women with OC, clinical investigation exploring the role of ruxolitinib in combination with chemotherapy agents or other targeted therapeutics is warranted.
Collapse
Affiliation(s)
- Irfan Yunianto
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
- Department of Biology Education, Universitas Ahmad Dahlan, Indonesia
| | - Margaret Currie
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
25
|
Morgan RD, Clamp AR, Barnes BM, Timms K, Schlecht H, Yarram-Smith L, Wallis Y, Valganon-Petrizan M, MacMahon S, White R, Morgan S, McKenna S, Hudson E, Tookman L, George A, Manchanda R, Sundar SS, Nicum S, Brenton JD, Kristeleit RS, Banerjee S, McNeish IA, Ledermann JA, Taylor SS, Evans DGR, Jayson GC. Homologous recombination deficiency in newly diagnosed FIGO stage III/IV high-grade epithelial ovarian cancer: a multi-national observational study. Int J Gynecol Cancer 2023; 33:1253-1259. [PMID: 37072323 DOI: 10.1136/ijgc-2022-004211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
OBJECTIVE Olaparib plus bevacizumab maintenance therapy improves survival outcomes in women with newly diagnosed, advanced, high-grade ovarian cancer with a deficiency in homologous recombination. We report data from the first year of routine homologous recombination deficiency testing in the National Health Service (NHS) in England, Wales, and Northern Ireland between April 2021 and April 2022. METHODS The Myriad myChoice companion diagnostic was used to test DNA extracted from formalin-fixed, paraffin-embedded tumor tissue in women with newly diagnosed International Federation of Gynecology and Obstetrics (FIGO) stage III/IV high-grade epithelial ovarian, fallopian tube, or primary peritoneal cancer. Tumors with homologous recombination deficiency were those with a BRCA1/2 mutation and/or a Genomic Instability Score (GIS) ≥42. Testing was coordinated by the NHS Genomic Laboratory Hub network. RESULTS The myChoice assay was performed on 2829 tumors. Of these, 2474 (87%) and 2178 (77%) successfully underwent BRCA1/2 and GIS testing, respectively. All complete and partial assay failures occurred due to low tumor cellularity and/or low tumor DNA yield. 385 tumors (16%) contained a BRCA1/2 mutation and 814 (37%) had a GIS ≥42. Tumors with a GIS ≥42 were more likely to be BRCA1/2 wild-type (n=510) than BRCA1/2 mutant (n=304). The distribution of GIS was bimodal, with BRCA1/2 mutant tumors having a higher mean score than BRCA1/2 wild-type tumors (61 vs 33, respectively, χ2 test p<0.0001). CONCLUSION This is the largest real-world evaluation of homologous recombination deficiency testing in newly diagnosed FIGO stage III/IV high-grade epithelial ovarian, fallopian tube, or primary peritoneal cancer. It is important to select tumor tissue with adequate tumor content and quality to reduce the risk of assay failure. The rapid uptake of testing across England, Wales, and Northern Ireland demonstrates the power of centralized NHS funding, center specialization, and the NHS Genomic Laboratory Hub network.
Collapse
Affiliation(s)
- Robert D Morgan
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew R Clamp
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Bethany M Barnes
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Helene Schlecht
- North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Yvonne Wallis
- Central and South Genomic Laboratory Hub, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Mikel Valganon-Petrizan
- North Thames Genomic Laboratory Hub, The Royal Marsden Hospital NHS Foundation Trust, Surrey, UK
| | - Suzanne MacMahon
- North Thames Genomic Laboratory Hub, The Royal Marsden Hospital NHS Foundation Trust, Surrey, UK
| | - Rhian White
- All Wales Genomics Laboratory, Institute of Medical Genetics, University Hospital Wales, Cardiff, UK
| | - Sian Morgan
- All Wales Genomics Laboratory, Institute of Medical Genetics, University Hospital Wales, Cardiff, UK
| | | | | | | | - Angela George
- The Royal Marsden NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Ranjit Manchanda
- Barts Health NHS Trust, London, UK
- Department of Health Services Research, The Faculty of Public Health and Policy, London School of Hygiene & Tropical Medicine, London, UK
- Wolfson Institute of Population Health, Queen Mary's University of London, London, UK
| | - Sudha S Sundar
- Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Shibani Nicum
- University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| | - James D Brenton
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Iain A McNeish
- Imperial College Healthcare NHS Trust, London, UK
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jonathan A Ledermann
- University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| | - Stephen S Taylor
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - D Gareth R Evans
- Manchester University NHS Foundation Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Gordon C Jayson
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
26
|
Yin M, Lu C, Zhou H, Liu Q, Yang J. Fibroblast Growth Factor 11 (FGF11) Promotes Progression and Cisplatin Resistance Through the HIF-1α/FGF11 Signaling Axis in Ovarian Clear Cell Carcinoma. Cancer Manag Res 2023; 15:753-763. [PMID: 37525667 PMCID: PMC10387280 DOI: 10.2147/cmar.s414703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/19/2023] [Indexed: 08/02/2023] Open
Abstract
Background A poor prognosis is often associated with ovarian clear cell carcinoma (OCCC) due to its relative resistance to platinum-based chemotherapy. Although several studies have been launched to explore the pathogenesis of OCCC, the mechanism of chemoresistance has yet to be uncovered. Methods Nanostring nCounter PanCancer Pathways Panel was performed to explore the expression profiles of OCCC tissues from patients showing different platinum sensitivity. Bioinformatic analysis was performed to select genes associated with chemoresistance and cell function assays, including colony formation, wound healing, transwell and flow cytometric analysis, were used to explore the role of the target gene in the progression of OCCC and resistance to cisplatin (DDP). Results Gene expression profiles and bioinformatic analysis verified that the expression of fibroblast growth factor 11 (FGF11) was significantly increased in platinum-resistant OCCC tissues and increased FGF11 expression was related to poorer survival. Downregulation of FGF11 inhibited the proliferation, migration, and invasion, reversing the DDP resistance of OCCC cells. Mechanically, FGF11 was regulated by hypoxia-inducible factor-1α (HIF-1α) to modulate the DDP sensitivity. Conclusion FGF11 was highly expressed in platinum-resistant OCCC tissues, promoting progression and resistance to DDP through the HIF-1α/FGF11 signaling axis.
Collapse
Affiliation(s)
- Min Yin
- National Clinical Research Center for Obstetric and Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Chunli Lu
- Neurospine Center, China International Neuroscience Institute (CHINA-INI), Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, People’s Republic of China
| | - Huimei Zhou
- National Clinical Research Center for Obstetric and Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Qian Liu
- National Clinical Research Center for Obstetric and Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jiaxin Yang
- National Clinical Research Center for Obstetric and Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
27
|
Batista MDP, Roffé M, Romero I, López-Guerrero JA, Illueca C, Lopez R, Balieiro Anastácio da Costa AA, De Brot L, Molina JP, Barboza L, Peria FM, Chaud F, Gouvêa Yamada AS, Poveda A, Rego EM. Genomic landscapes of ovarian clear cell carcinoma from latin countries reveal aberrations linked to survival and progression. BMC Cancer 2023; 23:613. [PMID: 37400764 DOI: 10.1186/s12885-023-11095-8] [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: 11/22/2022] [Accepted: 06/21/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Ovarian clear cell carcinomas (OCCCs) are rare, aggressive and chemoresistant tumors. Geographical and ethnic differences in the incidence of OCCC have been reported with a higher incidence in Asiatic countries. There is a paucity of information regarding OCCC in Latin America (LA) and other countries. METHODS Here, we characterized two cohorts of 33 patients with OCCC from LA (24 from Brazil and 9 from Costa Rica) and a cohort of 27 patients from Spain. Genomic analysis was performed for 26 OCCC using the OncoScan platform. Tumors were classified according to their genomic landscapes into subgroups. Clinical parameters were related to the frequency of genomic aberrations. RESULTS The median overall survival (OS) was not significantly different between the cohorts. Genomic landscapes were characterized by different homologous recombination deficiency (HRD) levels. No difference in the distribution of genomic landscapes profiles was detected between patients from the different cohorts. OCCCs with MYC-amplified tumors harboring a concomitant loss of a region in chromosome 13q12-q13 that includes the BRCA2 gene had the longest OS. In contrast, patients carrying a high number (> 30) of total copy number (CN) aberrations with no concomitant alterations in MYC and BRCA2 genes presented the shortest OS. Furthermore, amplification of the ASH1L gene was also associated with a shorter OS. Initial-stage OCCCs with early progression were characterized by gains in the JNK1 and MKL1 genes. CONCLUSIONS Our results provide new data from understudied OCCC populations and reveal new potential markers for OCCCs.
Collapse
Affiliation(s)
- Mariana de Paiva Batista
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil.
| | - Martín Roffé
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Ignacio Romero
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | | | - Carmen Illueca
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | - Raquel Lopez
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | | | - Louise De Brot
- Department of Pathology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Juan Pablo Molina
- Medical Oncology Service, México Hospital, CCSS, San José, Costa Rica
| | - Laura Barboza
- Pathological Anatomy Service, San Juan de Dios Hospital, San José, Costa Rica
| | - Fernanda Maris Peria
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Fernando Chaud
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Ana Silvia Gouvêa Yamada
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Andres Poveda
- Oncogynecologic Department, Quironsalud Hospital, Valencia, Spain
| | - Eduardo Magalhães Rego
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| |
Collapse
|
28
|
Quintela M, James DW, Garcia J, Edwards K, Margarit L, Das N, Lutchman-Singh K, Beynon AL, Rioja I, Prinjha RK, Harker NR, Gonzalez D, Steven Conlan R, Francis LW. In silico enhancer mining reveals SNS-032 and EHMT2 inhibitors as therapeutic candidates in high-grade serous ovarian cancer. Br J Cancer 2023; 129:163-174. [PMID: 37120667 PMCID: PMC10307814 DOI: 10.1038/s41416-023-02274-2] [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: 10/17/2022] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 05/01/2023] Open
Abstract
BACKGROUND Epigenomic dysregulation has been linked to solid tumour malignancies, including ovarian cancers. Profiling of re-programmed enhancer locations associated with disease has the potential to improve stratification and thus therapeutic choices. Ovarian cancers are subdivided into histological subtypes that have significant molecular and clinical differences, with high-grade serous carcinoma representing the most common and aggressive subtype. METHODS We interrogated the enhancer landscape(s) of normal ovary and subtype-specific ovarian cancer states using publicly available data. With an initial focus on H3K27ac histone mark, we developed a computational pipeline to predict drug compound activity based on epigenomic stratification. Lastly, we substantiated our predictions in vitro using patient-derived clinical samples and cell lines. RESULTS Using our in silico approach, we highlighted recurrent and privative enhancer landscapes and identified the differential enrichment of a total of 164 transcription factors involved in 201 protein complexes across the subtypes. We pinpointed SNS-032 and EHMT2 inhibitors BIX-01294 and UNC0646 as therapeutic candidates in high-grade serous carcinoma, as well as probed the efficacy of specific inhibitors in vitro. CONCLUSION Here, we report the first attempt to exploit ovarian cancer epigenomic landscapes for drug discovery. This computational pipeline holds enormous potential for translating epigenomic profiling into therapeutic leads.
Collapse
Affiliation(s)
- Marcos Quintela
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - David W James
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Jetzabel Garcia
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Kadie Edwards
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Lavinia Margarit
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
- Cwm Taf Morgannwg University Health Board, Swansea, SA2 8QA, UK
| | - Nagindra Das
- Swansea Bay University Health Board, Swansea, SA12 7BR, UK
| | | | | | - Inmaculada Rioja
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Rab K Prinjha
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Nicola R Harker
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Deyarina Gonzalez
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - R Steven Conlan
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Lewis W Francis
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK.
| |
Collapse
|
29
|
Ren J, Mao L, Zhao J, Li XL, Wang C, Liu XY, Jin ZY, He YL, Li Y, Xue HD. Seeing beyond the tumor: computed tomography image-based radiomic analysis helps identify ovarian clear cell carcinoma subtype in epithelial ovarian cancer. LA RADIOLOGIA MEDICA 2023:10.1007/s11547-023-01666-x. [PMID: 37368228 DOI: 10.1007/s11547-023-01666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
OBJECTIVE To develop and validate a model that can preoperatively identify the ovarian clear cell carcinoma (OCCC) subtype in epithelial ovarian cancer (EOC) using CT imaging radiomics and clinical data. MATERIAL AND METHODS We retrospectively analyzed data from 282 patients with EOC (training set = 225, testing set = 57) who underwent pre-surgery CT examinations. Patients were categorized into OCCC or other EOC subtypes based on postoperative pathology. Seven clinical characteristics (age, cancer antigen [CA]-125, CA-199, endometriosis, venous thromboembolism, hypercalcemia, stage) were collected. Primary tumors were manually delineated on portal venous-phase images, and 1218 radiomic features were extracted. The F-test-based feature selection method and logistic regression algorithm were used to build the radiomic signature, clinical model, and integrated model. To explore the effects of integrated model-assisted diagnosis, five radiologists independently interpreted images in the testing set and reevaluated cases two weeks later with knowledge of the integrated model's output. The diagnostic performances of the predictive models, radiologists, and radiologists aided by the integrated model were evaluated. RESULTS The integrated model containing the radiomic signature (constructed by four wavelet radiomic features) and three clinical characteristics (CA-125, endometriosis, and hypercalcinemia), showed better diagnostic performance (AUC = 0.863 [0.762-0.964]) than the clinical model (AUC = 0.792 [0.630-0.953], p = 0.295) and the radiomic signature alone (AUC = 0.781 [0.636-0.926], p = 0.185). The diagnostic sensitivities of the radiologists were significantly improved when using the integrated model (p = 0.023-0.041), while the specificities and accuracies were maintained (p = 0.074-1.000). CONCLUSION Our integrated model shows great potential to facilitate the early identification of the OCCC subtype in EOC, which may enhance subtype-specific therapy and clinical management.
Collapse
Affiliation(s)
- Jing Ren
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Li Mao
- AI Lab, Deepwise Healthcare, Beijing, People's Republic of China
| | - Jia Zhao
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Xiu-Li Li
- AI Lab, Deepwise Healthcare, Beijing, People's Republic of China
| | - Chen Wang
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Xin-Yu Liu
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Zheng-Yu Jin
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Yong-Lan He
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China.
| | - Yuan Li
- Department of Obstetrics and Gynecology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Beijing, People's Republic of China.
| | - Hua-Dan Xue
- Department of Radiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital, Shuai Fu Yuan 1, Dongcheng District, Beijing, 100730, People's Republic of China.
| |
Collapse
|
30
|
Sinsuwan W, Norchai P. Repurposing Drugs in Controlling Recurrent Platinum-Resistant Clear-Cell Ovarian Cancer. Case Rep Oncol Med 2023; 2023:2079654. [PMID: 37323625 PMCID: PMC10266906 DOI: 10.1155/2023/2079654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/11/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
Background Recurrent platinum-resistant clear-cell ovarian cancer has a low overall survival duration of 7-8 months, making it a fatal disease. Currently, chemotherapy is the major kind of treatment, but it offers little advantage. Repurposed conventional drugs have recently been found to offer the ability to control cancer with few side effects and at a reasonable cost to healthcare organizations. Case Presentation. In this case report, we present the case of a 41-year-old Thai female patient diagnosed with recurrent platinum-resistant clear-cell ovarian cancer (PRCCC) in the year 2020. After undergoing chemotherapy for two courses and failing to respond to treatment, she began alternative medicine with repurposing drugs in November 2020. Simvastatin, metformin, niclosamide, mebendazole, itraconazole, loratadine, and chloroquine were also administered. Two months after therapy, a computerized tomography (CT) scan revealed a conflict between a decline in tumor marker levels (CA 125, CA 19-9) and an increase in the number of lymph nodes. However, after continuing all medications for 4 months, the CA 125 level decreased from 303.6 to 54 U/ml, and the CA 19-9 level decreased from 1210.3 to 386.10 U/ml. The patient's EQ-5D-5L score increased from 0.631 to 0.829 (abdominal pain and depression), indicating improved quality of life. Overall survival was 8.5 months, and progression-free survival was 2 months. Conclusion The response to drug repurposing is demonstrated by a four-month-long improvement in symptoms. This work introduces a novel strategy for the management of recurrent platinum-resistant clear-cell ovarian cancer that needs further evaluation in large-scale studies.
Collapse
Affiliation(s)
- Woraporn Sinsuwan
- College of Integrative Medicine (CIM), Dhurakij Pundit University, Thailand
| | - Phawit Norchai
- College of Integrative Medicine (CIM), Dhurakij Pundit University, Thailand
| |
Collapse
|
31
|
Yin M, Lu C, Zhou H, Liu Q, Yang J. Differential molecular pathway expression according to chemotherapeutic response in ovarian clear cell carcinoma. BMC Womens Health 2023; 23:298. [PMID: 37270486 DOI: 10.1186/s12905-023-02420-1] [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: 02/13/2023] [Accepted: 05/09/2023] [Indexed: 06/05/2023] Open
Abstract
OBJECTIVE Ovarian clear cell carcinoma (OCCC) is a distinct entity from epithelial ovarian cancer. The prognosis of advanced and recurrent disease is very poor due to resistance to chemotherapeutic agents. Our aim was to explore the molecular alterations among OCCC patients with different chemotherapeutic responses and to obtain insights into potential biomarkers. METHODS Twenty-four OCCC patients were included in this study. The patients were divided into two groups based on the relapse time after the first-line platinum-based chemotherapy: the platinum-sensitive group (PS) and the platinum-resistant group (PR). Gene expression profiling was performed using NanoString nCounter PanCancer Pathways Panel. RESULTS Gene expression analysis comparing PR vs. PS identified 32 differentially expressed genes: 17 upregulated genes and 15 downregulated genes. Most of these genes are involved in the PI3K, MAPK and Cell Cycle-Apoptosis pathways. In particular, eight genes are involved in two or all three pathways. CONCLUSION The dysregulated genes in the PI3K, MAPK, and Cell Cycle-Apoptosis pathways identified and postulated mechanisms could help to probe biomarkers of OCCC platinum sensitivity, providing a research basis for further exploration of targeted therapy.
Collapse
Affiliation(s)
- Min Yin
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunli Lu
- Neurospine Center, Xuanwu Hospital, National Center for Neurological Disorders, China International Neuroscience Institute (CHINA-INI), Capital Medical University, Beijing, China
| | - Huimei Zhou
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qian Liu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
32
|
Long X, Lu H, Cai MC, Zang J, Zhang Z, Wu J, Liu X, Cheng L, Cheng J, Cheung LWT, Shen Z, Zhou Y, Di W, Zhuang G, Yin X. APOBEC3B stratifies ovarian clear cell carcinoma with distinct immunophenotype and prognosis. Br J Cancer 2023; 128:2054-2062. [PMID: 36997661 PMCID: PMC10206171 DOI: 10.1038/s41416-023-02239-5] [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: 04/18/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) is a challenging disease due to its intrinsic chemoresistance. Immunotherapy is an emerging treatment option but currently impeded by insufficient understanding of OCCC immunophenotypes and their molecular determinants. METHODS Whole-genome sequencing on 23 pathologically confirmed patients was employed to depict the genomic profile of primary OCCCs. APOBEC3B expression and digital pathology-based Immunoscore were assessed by performing immunohistochemistry and correlated with clinical outcomes. RESULTS An APOBEC-positive (APOBEC+) subtype was identified based on the characteristic mutational signature and prevalent kataegis events. APOBEC + OCCC displayed favourable prognosis across one internal and two external patient cohorts. The improved outcome was ascribable to increased lymphocytic infiltration. Similar phenomena of APOBEC3B expression and T-cell accumulation were observed in endometriotic tissues, suggesting that APOBEC-induced mutagenesis and immunogenicity could occur early during OCCC pathogenesis. Corroborating these results, a case report was presented for an APOBEC + patient demonstrating inflamed tumour microenvironment and clinical response to immune checkpoint blockade. CONCLUSIONS Our findings implicate APOBEC3B as a novel mechanism of OCCC stratification with prognostic value and as a potential predictive biomarker that may inform immunotherapeutic opportunities.
Collapse
Affiliation(s)
- Xiaoran Long
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaiwu Lu
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mei-Chun Cai
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyu Zang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuqing Zhang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Wu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoshi Liu
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Cheng
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiejun Cheng
- Department of Radiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lydia W T Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wen Di
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Guanglei Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xia Yin
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Lebreton C, Quesada S, Bini M, Babin G, Rossi L, Chopin N, Croce S, Hartog C, Renaud T, Gaillard AL, Petit A, Serre AA, Trédan O, Rowinski E, Cockenpot V, Treilleux I, Rousset-Jablonski C, Méeus P, Guyon F, Ray-Coquard I. [Treatments for rare ovarian tumors: What's new?]. Bull Cancer 2023:S0007-4551(23)00151-0. [PMID: 37045734 DOI: 10.1016/j.bulcan.2023.03.007] [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: 01/13/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/14/2023]
Abstract
Even if each rare ovarian tumor (ROT) has a low incidence, the sum of all these entities represents almost the half of all ovarian neoplasms. Thus, development of dedicated clinical trial emerged as a prerequisite to improve their managements. Owing to the spreading of dedicated institutional networks and (supra)national collaborations, the number of clinical trials has increased the past few years, with different types of trials; while some focused on specific molecular features, others assessed innovative molecules. Furthermore, relevant randomized clinical trials were designed as a mean to position new treatment options. Currently, innovative molecular-driven trials, based on master protocol trials are emerging and may shed light towards the improvement of personalized medicine regarding ROT.
Collapse
Affiliation(s)
- Coriolan Lebreton
- Institut Bergonié, département d'oncologie médicale, Bordeaux, France; University of Bordeaux, ARTiSt Lab, Inserm U1312, Bordeaux, France.
| | - Stanislas Quesada
- Institut régional du cancer de Montpellier, département d'oncologie médicale, Montpellier, France
| | - Marta Bini
- Instituto Nazionale dei Tumori, Milano, Italie
| | - Guillaume Babin
- University of Bordeaux, ARTiSt Lab, Inserm U1312, Bordeaux, France; Institut Bergonié, département de chirurgie, Bordeaux, France
| | - Léa Rossi
- Centre Léon-Bérard, département de chirurgie, Lyon, France
| | - Nicolas Chopin
- Centre Léon-Bérard, département de chirurgie, Lyon, France
| | - Sabrina Croce
- Institut Bergonié, département de biopathologie, Bordeaux, France; University of Bordeaux, SARCOTARGET Lab, Inserm U1312, Bordeaux, France
| | - Cécile Hartog
- University of Bordeaux, ARTiSt Lab, Inserm U1312, Bordeaux, France; Centre Léon-Bérard, département de chirurgie, Lyon, France
| | - Tiphaine Renaud
- Institut Bergonié, département de gynécologie médicale, Bordeaux, France
| | | | - Adeline Petit
- Institut Bergonié, département de radiothérapie et curiethérapie, Bordeaux, France
| | - Anne-Agathe Serre
- Centre Léon-Bérard, département de radiothérapie et curiethérapie, Lyon, France
| | - Olivier Trédan
- Centre Léon-Bérard, département d'oncologie médicale, Lyon, France
| | - Elise Rowinski
- Centre Léon-Bérard, département d'oncologie médicale, Lyon, France
| | | | | | | | - Pierre Méeus
- Centre Léon-Bérard, département de chirurgie, Lyon, France
| | - Frédéric Guyon
- Institut Bergonié, département de chirurgie, Bordeaux, France
| | - Isabelle Ray-Coquard
- Centre Léon-Bérard, département d'oncologie médicale, Lyon, France; University of Claude-Bernard Lyon 1, health services and performance research lab (EA 7425 HESPER), Lyon, France
| |
Collapse
|
34
|
Sun M, Jiang W. Ovarian clear cell carcinoma with or without endometriosis origin in a single institution cohort. Discov Oncol 2023; 14:39. [PMID: 37004660 PMCID: PMC10067778 DOI: 10.1007/s12672-023-00649-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND As ovarian clear cell carcinoma (OCCC) has distinct clinical features, biology, genetic characteristics and mechanisms of pathogenesis, and whether the origin of endometriosis or not affects the prognosis of OCCC remains controversial. METHODS We retrospectively collected medical records and follow-up data of patients with OCCC treated at the Obstetrics and Gynecology Hospital of Fudan University from January 2009 to December 2019. Further, we divided patients into 2 groups. Group 1: non-endometriosis origin; Group 2: endometriosis origin. Clinicopathological characteristics and survival outcomes were compared between the 2 groups. RESULTS A total of one hundred and twenty-five patients with ovarian clear cell carcinoma were identified and included. In the overall patients' population, the 5 year overall survival was 84.8%, the mean overall survival was 85.9 months. The results of the stratified analysis showed that early stage (FIGO stage I/II) OCCC had a good prognosis. The results of univariate analyses indicated that a statistically significant relationship between overall survival (OS) and FIGO stage, lymph node metastasis, peritoneum metastasis, chemotherapy administration methods, Chinese herbal treatment, molecular target therapy. As for progression-free survival (PFS), a significant relationship between PFS and child-bearing history, largest residual tumor size, FIGO stage, tumor maximum diameter, lymph node metastasis was found, respectively. FIGO stage and lymph node metastasis are common poor prognostic factors affecting OS and PFS. The multivariate regression analysis revealed that FIGO stage (p = 0.028; HR, 1.944; 95% CI 1.073-3.52) and treatment by Chinese herbs (p = 0.018; HR, 0.141; 95% CI 0.028-0.716) were identified as influencing factors with regard to survival. The presence or absence of lymphadenectomy did not affect OS of 125 OCCC patients (p = 0.851; HR, 0.825; 95% CI 0.111-6.153). There was a trend towards a better prognosis for patients with OCCC of endometriosis origin than those with OCCC of non-endometriosis origin (p = 0.062; HR, 0.432; 95% CI 0.179-1.045). The two groups differed with respect to several clinicopathological factors. And the proportion of patients with disease relapse was higher in Group 1 (46.9%) than in Group 2 (25.0%), with a statistically significant difference (p = 0.048). CONCLUSIONS Surgical staging and treatment by Chinese herbs postoperatively are two independent prognostic factors affecting the OS of OCCC, early detection and Chinese herbal medicine combined with chemotherapy postoperatively may be a good choice. Tumor with endometriosis-origin was found less likely to relapse. While the non-necessity of lymphadenectomy in advanced ovarian cancer has been proven, the need for lymphadenectomy in the early stage ovarian cancer, including early stage OCCC, still deserved to be explored.
Collapse
Affiliation(s)
- Mingming Sun
- Department Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, People's Republic of China
| | - Wei Jiang
- Department Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, People's Republic of China.
| |
Collapse
|
35
|
Quesada S, Bini M, Lebreton C, Ray-Coquard I. Update on new treatments for rare ovarian tumours. Curr Opin Obstet Gynecol 2023; 35:27-33. [PMID: 36440753 PMCID: PMC9788923 DOI: 10.1097/gco.0000000000000836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW In spite of their rarity when considered individually, the sum of all rare ovarian tumours (ROT) represent almost half of all ovarian malignancies. As such, their appropriate inclusion within dedicated clinical trials is essential for enhanced management. RECENT FINDINGS Supported by institutional expert national (e.g. TMRG) and international (e.g. ESGO) networks and owing to national (e.g. ARCAGY-GINECO) and international (e.g. ENGOT) collaborations dedicated to clinical research, the last few years have shown increased number of clinical trials dedicated to ROT. These either were based on specific molecular features of ROT (e.g. expression of oestrogen receptors for low-grade serous ovarian carcinomas and anastrazole evaluation in the PARAGON trial) or on the evaluation of innovative therapies (e.g. pembrolizumab within the ROT cohort from the AcSé Pembrolizumab multicentric basket trial). Furthermore, recent years have also shown the advent of randomized clinical trials. For instance, the ALIENOR trial positioned weekly paclitaxel as a new option for relapsed sex cord-stromal tumours, while the GOG281/LOGS trial raised trametinib as a new standard-of-care option for recurrent low-grade serous carcinomas. SUMMARY The last few years have exhibited a paradigm shift towards the possibility to develop dedicated trials for ROT, owing to international collaborations supported by institutional networks. Current trials, molecular-driven and based on innovative designs, are highly promising, as they may bring ROT management towards more personalized medicine.
Collapse
Affiliation(s)
- Stanislas Quesada
- Centre Léon Bérard, Lyon, France
- Institut régional du Cancer de Montpellier, Montpellier, France
| | - Marta Bini
- Centre Léon Bérard, Lyon, France
- Instituto Nazionale dei Tumori, Milan, Italy
| | | | | |
Collapse
|
36
|
Wang CK, Chen TJ, Tan GY, Chang FP, Sridharan S, Yu CHA, Chang YH, Chen YJ, Cheng LT, Hwang-Verslues WW. MEX3A Mediates p53 Degradation to Suppress Ferroptosis and Facilitate Ovarian Cancer Tumorigenesis. Cancer Res 2023; 83:251-263. [PMID: 36354374 PMCID: PMC9845988 DOI: 10.1158/0008-5472.can-22-1159] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/24/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022]
Abstract
Epithelial ovarian cancer is a highly heterogeneous and malignant female cancer with an overall low survival rate. Mutations in p53 are prevalent in the major ovarian cancer histotype, high-grade serous ovarian carcinoma (HGSOC), while p53 mutations are much less frequent in other ovarian cancer subtypes, particularly in ovarian clear cell carcinoma (OCCC). Advanced stage OCCC with wild-type (WT) p53 has a worse prognosis and increased drug resistance, metastasis, and recurrence than HGSOC. The mechanisms responsible for driving the aggressiveness of WT p53-expressing ovarian cancer remain poorly understood. Here, we found that upregulation of MEX3A, a dual-function protein containing a RING finger domain and an RNA-binding domain, was critical for tumorigenesis in WT p53-expressing ovarian cancer. MEX3A overexpression enhanced the growth and clonogenicity of OCCC cell lines. In contrast, depletion of MEX3A in OCCC cells, as well as ovarian teratocarcinoma cells, reduced cell survival and proliferative ability. MEX3A depletion also inhibited tumor growth and prolonged survival in orthotopic xenograft models. MEX3A depletion did not alter p53 mRNA level but did increase p53 protein stability. MEX3A-mediated p53 protein degradation was crucial to suppress ferroptosis and enhance tumorigenesis. Consistently, p53 knockdown reversed the effects of MEX3A depletion. Together, our observations identified MEX3A as an important oncogenic factor promoting tumorigenesis in ovarian cancer cells expressing WT p53. SIGNIFICANCE Degradation of p53 mediated by MEX3A drives ovarian cancer growth by circumventing p53 tumor suppressive functions, suggesting targeting MEX3A as a potential strategy for treating of ovarian cancer expressing WT p53.
Collapse
Affiliation(s)
- Cheng-Kai Wang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Tzu-Jou Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Grace Y.T. Tan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Fang-Pei Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | - Yen-Hou Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Jen Chen
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Tzu Cheng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wendy W. Hwang-Verslues
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Corresponding Author: Wendy W. Hwang-Verslues, Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 115, Taiwan. Phone: +886-2-2787-1246; Fax: +886-2-2789-9924; E-mail:
| |
Collapse
|
37
|
Angelina YA, Tjokroprawiro BA. Advanced Stage Clear Cell Ovarian Carcinoma Mimicking Uterine Sarcoma Without Gross Residual Tumor During Primary Surgery: A Case Report. CLINICAL MEDICINE INSIGHTS-CASE REPORTS 2023; 16:11795476231166623. [PMID: 37056468 PMCID: PMC10088404 DOI: 10.1177/11795476231166623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/13/2023] [Indexed: 04/08/2023]
Abstract
Background: Clear cell ovarian carcinoma is rare and accounts for 1%-12% of ovarian epithelial carcinomas, depending on ethnicity. The prevalence of clear cell ovarian carcinoma in Asian, White, and Black women is 11.1%%, 4.8%, and 3.1%, respectively. Magnetic resonance imaging (MRI) shows that clear cell ovarian carcinomas are typically unilocular cyst-solid (34.9%) or multilocular-solid (41.4%); only 23.7% are solid with papillary projections. MRI can detect clear cell ovarian carcinoma with a sensitivity and specificity of 90% and 87%, respectively. Notably, sometimes ovarian masses have a solid feature and should be differentiated from uterine masses. Clear cell ovarian carcinoma has a better prognosis compared to serous carcinoma when diagnosed at an early stage, but it has a poorer prognosis at an advanced stage. The absence of a residual tumor is a favorable prognostic factor in patients with advanced-stage clear cell ovarian carcinoma. Herein, we present a case in which clear cell ovarian carcinoma was misdiagnosed as uterine sarcoma because imaging showed a mass with a solid uterine-like and necrotic area. In the present case, cytoreductive surgery was performed to remove the entire tumor and its infiltration to the sigmoid colon and left ureter. Hence, the patient had a better prognosis. Case report: A 57-year-old Indonesian woman presented to our hospital (Dr. Soetomo General Hospital) with post-menopausal bleeding, a large solid pelvic mass, and abdominal discomfort. The patient was diagnosed with uterine sarcoma due to the solid feature observed during ultrasonography and MRI. During the surgery, the mass was observed to originate from the left ovary, and primary debulking surgery with a multidisciplinary team was performed with zero residual tumor tissue. The tumor was histopathologically confirmed as clear cell carcinoma. Conclusion: MRI of clear cell ovarian carcinoma can be misdiagnosed as uterine sarcoma due to its solid feature. Additionally, the enlarged mass distorts the anatomical landmarks. Surgery with no residual tumor improves the prognosis for advanced-stage clear cell ovarian carcinoma.
Collapse
Affiliation(s)
- Yoan Alexandria Angelina
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga/Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Brahmana Askandar Tjokroprawiro
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga/Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
38
|
Ghasemi D, Ameli F, Nili F, Edjtemaei R, Sheikhhasani S. Immunohistochemical expression of PD-L1 and its correlation with microsatellite status in endometrial and ovarian clear cell carcinomas: a cross-sectional study. BMC Cancer 2022; 22:1362. [PMID: 36581850 PMCID: PMC9801577 DOI: 10.1186/s12885-022-10478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Clear cell carcinoma is an uncommon histologic subtype of ovarian and endometrial carcinoma with poor response to Platinium-based chemotherapy agents at high stages. Blockage of Programmed cell Death Ligand-1 (PD-L1), can be used in targeted immunotherapy. This study investigated Mismatch Repair Deficiency (MMR-D) status, PD-L1 expression, and the correlation between PD-L1 expression and microsatellite instability (MSI) status in ovarian and endometrial clear cell carcinomas. METHODS Ovarian clear cell carcinoma (OCCC) (n = 28) and endometrial clear cell carcinoma (ECCC) (n = 28) samples were evaluated for PD-L1 (in tumoral and peri-tumoral inflammatory cells), MSH6 and PMS2 expression by immunohistochemistry (IHC) study. PD-L1 expression > 1% in tumor cells and > 5% in peritumoral inflammatory cells were considered positive. RESULTS The prevalence of PD-L1 expression was higher in ECCC (20/28, 71.43%) compared to OCCC tumor cells (16/28, 57.15%) (p > 0.05), while expression in peritumoral inflammatory cells was significantly higher in ECCC (25/28, 89.29%) compared to OCCC (11/28, 39.28%) (p < 0.05). MMR-D was observed in 5 cases, four OCCCs and one ECCC, among which, four (80%) showed PD-L1 expression in peritumoral inflammatory and tumor cells. The only OCCC case with extensive PD-L1 expression in tumor cells (> 50%) exhibited MSH6/MSH2 loss. No significant correlation was noted between PD-L1 expression and the pathologic stage or survival. CONCLUSION PD-L1 expression was significantly associated with clear cell morphology, especially in the endometrium, independent of MMR protein status.
Collapse
Affiliation(s)
- Dorsa Ghasemi
- grid.414574.70000 0004 0369 3463Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, End of Keshavarz Ave, Tehran, IR Iran
| | - Fereshteh Ameli
- grid.414574.70000 0004 0369 3463Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, End of Keshavarz Ave, Tehran, IR Iran
| | - Fatemeh Nili
- grid.414574.70000 0004 0369 3463Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, End of Keshavarz Ave, Tehran, IR Iran
| | - Ramtin Edjtemaei
- grid.414574.70000 0004 0369 3463Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, End of Keshavarz Ave, Tehran, IR Iran
| | - Shahrzad Sheikhhasani
- grid.414574.70000 0004 0369 3463Department of Gynecology Oncology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, IR Iran
| |
Collapse
|
39
|
Phase II study of gemcitabine, cisplatin, and bevacizumab for first recurrent and refractory ovarian clear cell carcinoma Kansai Clinical Oncology Group-G1601. Anticancer Drugs 2022:00001813-990000000-00138. [PMID: 36729915 DOI: 10.1097/cad.0000000000001472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Patients with advanced ovarian clear cell carcinoma (CCC) have a poor prognosis in the absence of an effective standard treatment. Combination therapy with gemcitabine, cisplatin, and bevacizumab (GPBev) is promising for ovarian CCC. Thus, we conducted a multi-institutional, phase II trial in Japan to examine the efficacy and safety of GPBev for CCC. This is the first study on the use of GPBev for CCC. Eighteen patients (median age, 56.5 years) with pathologically confirmed first recurrent or refractory CCC and having evaluable regions, as assessed using RECIST, were recruited between January 2017 and May 2019. Gemcitabine (1000 mg/m2), cisplatin (40 mg/m2), and bevacizumab (10 mg/kg) were administered intravenously on days 1 and 15, every 28 days, for 6-10 cycles, until disease progression or intolerable toxicity. The primary endpoint was overall response rate (ORR). The secondary endpoints included disease control rate (DCR) and adverse events (AEs). Fifteen patients (83.3%) completed 6-10 cycles of treatment; three patients (two with AEs and one with progressive disease) did not. The ORR was 61.1% [complete response (CR) 3 and partial response (PR) 8] and DCR was 88.9% (CR 3, PR 8, and stable disease 5). Grade 3 and 4 hematological AEs were observed in 16.7 and 5.6% of the patients, respectively. Nonhematological AEs of grades 3 and 4 were observed in 27.8 and 5.6% of the patients, respectively. GPBev is a promising therapy for CCC owing to the high ORR and acceptable toxicity for the first recurrence and refractory CCC.
Collapse
|
40
|
Manning-Geist B, Gordhandas S, Hodgson A, Zhou QC, Iasonos A, Chi DS, Ellenson L, Aghajanian CA, Abu-Rustum NR, Leitao M, Long K, Rubinstein MM, Sonoda Y, Alektiar K, Weigelt B, Zivanovic O, Grisham RN. Management of patients with early-stage ovarian clear cell carcinoma: risk stratification and fertility conservation. Int J Gynecol Cancer 2022; 32:1576-1582. [PMID: 36368707 PMCID: PMC9822864 DOI: 10.1136/ijgc-2022-003935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We sought to describe clinicopathologic and treatment factors associated with oncologic outcomes in patients with early-stage ovarian clear cell carcinoma undergoing complete staging and in a sub-set of these patients undergoing fertility-conserving surgery. METHODS We retrospectively identified patients with ovarian clear cell carcinoma initially treated at our institution from January 1, 1996 to March 31, 2020. Survival was estimated using Kaplan-Meier curves and compared by log-rank test. Survival-associated variables were identified by Cox proportional hazards regression. RESULTS Of 182 patients, mismatch repair and p53 protein expression were assessed by immunohistochemistry on 82 and 66 samples, respectively. There were no significant differences in progression-free survival or overall survival between mismatch repair-deficient (n=6, including 4 patients with Lynch syndrome; 7.3%) and mismatch repair-proficient patients, whereas aberrant p53 expression (n=3; 4.5%) was associated with worse progression-free (p<0.001) and overall survival (p=0.01). Patients with stage IA/IC1 disease had a 95% 5-year overall survival rate (95% CI 88% to 98%); patients with stage IC2/IC3 disease had a similar 5-year overall survival rate (76%; 95% CI 54% to 88%) to that of patients with stage IIA/IIB disease (82%; 95% CI 54% to 94%). There was no difference in 5-year overall survival in patients with stage IA/IC1 undergoing chemotherapy versus observation (94% vs 100%). Nine patients underwent fertility-sparing surgery and none experienced recurrence. Of five patients who pursued fertility, all had successful pregnancies. CONCLUSIONS In patients with completely staged ovarian clear cell carcinoma, those with stage IA/IC1 disease have an excellent prognosis, regardless of chemotherapy. Aberrant p53 expression may portend worse outcomes. Additional investigation is warranted on the safety of fertility conservation in patients with stage IA/IC1 disease.
Collapse
Affiliation(s)
- Beryl Manning-Geist
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sushmita Gordhandas
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anjelica Hodgson
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Qin C Zhou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexia Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dennis S Chi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lora Ellenson
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Carol A Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nadeem R Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mario Leitao
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kara Long
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria M Rubinstein
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yukio Sonoda
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kaled Alektiar
- Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Oliver Zivanovic
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Rachel N Grisham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
41
|
Purwar R, Ranjan R, Soni K, Pandey M, Upadhyay SK, Pai E, Kumar T. Lymphadenectomy in ovarian cancers: a meta-analysis of hazard ratios from randomized clinical trials. World J Surg Oncol 2022; 20:367. [PMID: 36419077 PMCID: PMC9682714 DOI: 10.1186/s12957-022-02835-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The debate surrounding systematic lymphadenectomy in the epithelial cancers of the ovary (EOC) was temporarily put to rest by the LION trial. However, there was a glaring disparity between the number of patients registered and the number of patients randomized suggesting inadvertent selection. A subsequent meta-analysis after this trial included all types of studies in the literature (randomized, non-randomized, case series, and, retrospective cohort), thus diluting the results. METHODS We conducted a meta-analysis of hazard ratios of randomized controlled trials, to study the role of systematic para-aortic and pelvic lymph node dissection in the EOC. A detailed search of MEDLINE, Cochrane, and Embase databases was done to look for the published randomized controlled trials (RCT) comparing lymphadenectomy versus no lymphadenectomy in EOC. A meta-analysis of hazard ratios (HR) was performed for overall survival (OS) and progression-free survival (PFS) using fixed and random effect models. The quality of the RCTs was evaluated on Jadad's score, and the risk of bias was estimated by the Cochrane tool. RESULTS A total of 1342 patients with EOC were included for quantitative analysis. On meta-analysis, HR for PFS was 0.9 (95% CI 0.79-1.04) favoring lymphadenectomy. HR for OS was 1 (95% CI 0.84-1.18) signifying no benefit of systematic lymphadenectomy. CONCLUSION The results show a trend towards increased PFS which did not reach statistical significance nor translate into any meaningful benefit in OS. There is still a need for a greater number of well-conducted, suitably powered trials to convincingly answer this question.
Collapse
Affiliation(s)
- Roli Purwar
- grid.463154.10000 0004 1768 1906Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| | - Rakesh Ranjan
- grid.411507.60000 0001 2287 8816Department of Science and Technology (DST), Centre for Interdisciplinary Mathematical Sciences, Banaras Hindu University, Varanasi, 221005 India
| | - Kishan Soni
- grid.463154.10000 0004 1768 1906Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| | - Manoj Pandey
- grid.463154.10000 0004 1768 1906Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| | - Satyanshu K. Upadhyay
- grid.411507.60000 0001 2287 8816Department of Statistics, Banaras Hindu University, Varanasi, 221005 India
| | - Esha Pai
- Department of Surgical Oncology, Heritage Hospitals, Varanasi, 221005 India
| | - Tarun Kumar
- grid.463154.10000 0004 1768 1906Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
42
|
Circular RNAs in Epithelial Ovarian Cancer: From Biomarkers to Therapeutic Targets. Cancers (Basel) 2022; 14:cancers14225711. [PMID: 36428803 PMCID: PMC9688053 DOI: 10.3390/cancers14225711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer, and more than 70% of patients are diagnosed at advanced stages. Despite the application of surgery and chemotherapy, the prognosis remains poor due to the high relapse rate. It is urgent to identify novel biomarkers and develop novel therapeutic strategies for EOC. Circular RNAs (circRNAs) are a class of noncoding RNAs generated from the "back-splicing" of precursor mRNA. CircRNAs exert their functions via several mechanisms, including acting as miRNA sponges, interacting with proteins, regulating transcription, and encoding functional proteins. Recent studies have identified many circRNAs that are dysregulated in EOC and may be used as diagnostic and prognostic markers. Increasing evidence has revealed that circRNAs play a critical role in ovarian cancer progression by regulating various cellular processes, including proliferation, apoptosis, metastasis, and chemosensitivity. The circRNA-based therapy may be a novel strategy that is worth exploring in the future. Here, we provide an overview of EOC and circRNA biogenesis and functions. We then discuss the dysregulations of circRNAs in EOC and the possibility of using them as diagnostic/prognostic markers. We also summarize the role of circRNAs in regulating ovarian cancer development and speculate their potential as therapeutic targets.
Collapse
|
43
|
Bolton KL, Chen D, Corona de la Fuente R, Fu Z, Murali R, Köbel M, Tazi Y, Cunningham JM, Chan IC, Wiley BJ, Moukarzel LA, Winham SJ, Armasu SM, Lester J, Elishaev E, Laslavic A, Kennedy CJ, Piskorz A, Sekowska M, Brand AH, Chiew YE, Pharoah P, Elias KM, Drapkin R, Churchman M, Gourley C, DeFazio A, Karlan B, Brenton JD, Weigelt B, Anglesio MS, Huntsman D, Gayther S, Konner J, Modugno F, Lawrenson K, Goode EL, Papaemmanuil E. Molecular Subclasses of Clear Cell Ovarian Carcinoma and Their Impact on Disease Behavior and Outcomes. Clin Cancer Res 2022; 28:4947-4956. [PMID: 35816189 PMCID: PMC9777703 DOI: 10.1158/1078-0432.ccr-21-3817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/24/2022] [Accepted: 07/07/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE To identify molecular subclasses of clear cell ovarian carcinoma (CCOC) and assess their impact on clinical presentation and outcomes. EXPERIMENTAL DESIGN We profiled 421 primary CCOCs that passed quality control using a targeted deep sequencing panel of 163 putative CCOC driver genes and whole transcriptome sequencing of 211 of these tumors. Molecularly defined subgroups were identified and tested for association with clinical characteristics and overall survival. RESULTS We detected a putative somatic driver mutation in at least one candidate gene in 95% (401/421) of CCOC tumors including ARID1A (in 49% of tumors), PIK3CA (49%), TERT (20%), and TP53 (16%). Clustering of cancer driver mutations and RNA expression converged upon two distinct subclasses of CCOC. The first was dominated by ARID1A-mutated tumors with enriched expression of canonical CCOC genes and markers of platinum resistance; the second was largely comprised of tumors with TP53 mutations and enriched for the expression of genes involved in extracellular matrix organization and mesenchymal differentiation. Compared with the ARID1A-mutated group, women with TP53-mutated tumors were more likely to have advanced-stage disease, no antecedent history of endometriosis, and poorer survival, driven by their advanced stage at presentation. In women with ARID1A-mutated tumors, there was a trend toward a lower rate of response to first-line platinum-based therapy. CONCLUSIONS Our study suggests that CCOC consists of two distinct molecular subclasses with distinct clinical presentation and outcomes, with potential relevance to both traditional and experimental therapy responsiveness. See related commentary by Lheureux, p. 4838.
Collapse
Affiliation(s)
- Kelly L. Bolton
- Washington University School of Medicine, St. Louis, Missouri
| | - Denise Chen
- Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | | | - Zhuxuan Fu
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | | | - Martin Köbel
- The University of Calgary, Calgary, Alberta, Canada
| | - Yanis Tazi
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | - Brian J. Wiley
- Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | - Jenny Lester
- David Geffen School of Medicine, Department of Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, California
| | - Esther Elishaev
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Angela Laslavic
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Catherine J. Kennedy
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
| | - Anna Piskorz
- University of Cambridge, Cambridge, United Kingdom
| | | | - Alison H. Brand
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
- The University of Sydney, Sydney, New South Wales, Australia
| | - Yoke-Eng Chiew
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
| | - Paul Pharoah
- University of Cambridge, Cambridge, United Kingdom
| | | | - Ronny Drapkin
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Anna DeFazio
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
- The 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
| | - Beth Karlan
- David Geffen School of Medicine, Department of Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, California
| | | | - Britta Weigelt
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - David Huntsman
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Simon Gayther
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Jason Konner
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Francesmary Modugno
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
44
|
Abstract
The RAS family of proteins is among the most frequently mutated genes in human malignancies. In ovarian cancer (OC), the most lethal gynecological malignancy, RAS, especially KRAS mutational status at codons 12, 13, and 61, ranges from 6-65% spanning different histo-types. Normally RAS regulates several signaling pathways involved in a myriad of cellular signaling cascades mediating numerous cellular processes like cell proliferation, differentiation, invasion, and death. Aberrant activation of RAS leads to uncontrolled induction of several downstream signaling pathways such as RAF-1/MAPK (mitogen-activated protein kinase), PI3K phosphoinositide-3 kinase (PI3K)/AKT, RalGEFs, Rac/Rho, BRAF (v-Raf murine sarcoma viral oncogene homolog B), MEK1 (mitogen-activated protein kinase kinase 1), ERK (extracellular signal-regulated kinase), PKB (protein kinase B) and PKC (protein kinase C) involved in cell proliferation as well as maintenance pathways thereby driving tumorigenesis and cancer cell propagation. KRAS mutation is also known to be a biomarker for poor outcome and chemoresistance in OC. As a malignancy with several histotypes showing varying histopathological characteristics, we focus on reviewing recent literature showcasing the involvement of oncogenic RAS in mediating carcinogenesis and chemoresistance in OC and its subtypes.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anjana Anand
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| | | | | | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Shahab Uddin
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| |
Collapse
|
45
|
Yuwono NL, Alonso A, Abbott J, Houshdaran S, Henry CE, Rodgers R, Ford CE, Warton K. Circulating cell-free endometrial DNA level is unaltered during menstruation and in endometriosis. Hum Reprod 2022; 37:2560-2569. [PMID: 36166696 DOI: 10.1093/humrep/deac198] [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: 01/13/2022] [Revised: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Is circulating cell-free DNA (cirDNA) from the endometrium elevated during menstruation and in endometriosis? SUMMARY ANSWER Endometrial cirDNA does not increase during menstruation and is not elevated in endometriosis. WHAT IS KNOWN ALREADY Changes in cirDNA associated with common benign conditions are a potential source of false positives in cancer diagnostic applications, but also present an opportunity for biomarker development for diseases such as endometriosis. Elevated cirDNA has been reported in endometriosis patients compared to healthy community controls, but no difference in total or endometrial cirDNA has been found between patients with endometriosis and patients with other gynaecological conditions. Likewise, menstruation is a potential driver of changes in cirDNA levels and tissue profile, but total and endothelial cirDNA do not increase during menstruation. STUDY DESIGN, SIZE, DURATION For endometriosis comparisons, 59 participants with surgically confirmed endometriosis and 27 laparoscopic patients without endometriosis (hospital controls) were prospectively recruited, while 25 healthy community participants (healthy controls) were recruited in a university setting. Total and endometrial cirDNA and cirDNA fragmentation were measured across the three groups. For menstrual comparisons, 36 matched non-menstruating and menstruating samples were collected from healthy women recruited within a university setting, and the endometrial cirDNA was compared between the two groups. PARTICIPANTS/MATERIALS, SETTING, METHODS cirDNA was extracted from venous blood plasma then quantitated by quantitative PCR of ALU repetitive element (115 bp) and TP53 gene sequence (105 bp) for total concentration. cirDNA derived from the endometrium was quantitated by methylation-specific droplet digital PCR of a FAM101A region (69 bp) after bisulfite conversion of the DNA. A cirDNA size fragmentation ratio was obtained by quantifying a long segment of ALU repetitive element (247 bp) and expressing the amount relative to the 115 bp ALU target. MAIN RESULTS AND THE ROLE OF CHANCE No differences in cirDNA level were found in any comparison populations in this study. Mean total cirDNA was unchanged between healthy controls (ALU-115-3.31 ng/ml; TP53-2.73 ng/ml), hospital controls (ALU-115-3.47 ng/ml; TP53-2.83 ng/ml) and endometriosis patients (ALU-115-3.35 ng/ml; TP53-2.66 ng/ml). Likewise, endometrial cirDNA was unchanged between healthy controls (18.3 copies/ml), hospital controls (20.6 copies/ml) and endometriosis patients (22 copies/ml). Endometrial cirDNA did not change during menstruation (non-menstruating: 38 copies/ml; menstruating: 33 copies/ml). Irrespective of endometriosis diagnosis, blood from patients undergoing laparoscopy (hospital controls: 0.77; endometriosis patients: 0.79), had a significantly higher cirDNA size ratio than community-recruited healthy controls (0.64), indicating increased abundance of long cirDNA fragments. LIMITATIONS, REASONS FOR CAUTION It was not possible to completely match the age, BMI and parity between the three cohorts investigated, however of these, only age has been shown to influence circulating DNA levels and not within the age range of our cohort. Blood from community-recruited healthy women and women undergoing laparoscopy was collected via antecubital vein venepuncture (processed within 3 h) and with either peripheral cannula or venepuncture (processed within 6 h), respectively, which could potentially impact the size distribution of circulating DNA fragments. For the collection of non-menstruating phase blood samples, we did not differentiate between follicular phase, ovulation and luteal phase. Thus, only the mensturating samples were collected at a consistent phase, and any fluctuations in cirDNA that occur at the other phases may have obscured small changes during menstruation. WIDER IMPLICATIONS OF THE FINDINGS There is no evidence that cirDNA has potential as a diagnostic biomarker for endometriosis. Endometriosis, representing a common benign gynaecological condition, and menstruation, representing a normal physiological occurrence in women, should not affect methylation-based diagnostics in other disease areas, including oncology. STUDY FUNDING/COMPETING INTEREST(S) N.L.Y.: Australian Government Research Training Program (RTP) Stipend through The University of New South Wales, Translational Cancer Research Network PhD Scholarship Top-Up Award via the Cancer Institute NSW, Beth Yarrow Memorial Award in Medical Science, UNSW Completion Scholarship; C.E.H.: Gynaecological Oncology Fund of the Royal Hospital for Women; K.W.: Ovarian Cancer Research Foundation and CAMILLA AND MARC. C.E.F.: UNSW Women's Wellbeing Academy and the Australian Human Rights Institute. We declare the following competing interest: K.W. holds stock in Guardant Health, Exact Sciences and Epigenomics AG. No other authors have competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- N L Yuwono
- Department of Obstetrics and Gynaecology, Gynaecological Cancer Research Group, Adult Cancer Program, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - A Alonso
- Department of Obstetrics and Gynaecology, Gynaecological Research and Clinical Evaluation (GRACE) Unit, Royal Hospital for Women, Sydney, NSW, Australia.,Department of Obstetrics and Gynaecology, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - J Abbott
- Department of Obstetrics and Gynaecology, Gynaecological Research and Clinical Evaluation (GRACE) Unit, Royal Hospital for Women, Sydney, NSW, Australia.,Department of Obstetrics and Gynaecology, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - S Houshdaran
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - C E Henry
- Department of Obstetrics and Gynaecology, Gynaecological Cancer Research Group, Adult Cancer Program, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - R Rodgers
- Department of Obstetrics and Gynaecology, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.,Department of Gynaecology and Reproductive Medicine, Royal Hospital for Women, Sydney, NSW, Australia
| | - C E Ford
- Department of Obstetrics and Gynaecology, Gynaecological Cancer Research Group, Adult Cancer Program, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - K Warton
- Department of Obstetrics and Gynaecology, Gynaecological Cancer Research Group, Adult Cancer Program, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
46
|
Farah AM, Gu S, Jia Y. Clinical analysis and literature review of a case of ovarian clear cell carcinoma with PIK3CA gene mutation: A case report. Medicine (Baltimore) 2022; 101:e30666. [PMID: 36123851 PMCID: PMC9478318 DOI: 10.1097/md.0000000000030666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Ovarian clear cell carcinoma (OCCC) is an uncommon malignant form of 5 subtypes of ovarian cancer, accounting for approximately 5% to 25% of all ovarian cancers. OCCC is usually diagnosed at a young age and an early stage. More than 50% of patients are associated with endometriosis. It shows less sensitivity to platinum-based chemotherapies, high recurrence, and poor prognosis, especially late. However, platinum-based chemotherapies remain the first-line treatment. Meanwhile, new treatment modalities have been explored, including immune checkpoint inhibitors and PI3K-AKT-mTOR pathway inhibitors. PATIENT CONCERN A 48-year-old Chinese woman, Gravida2 Para1, complained of irregular and painful vaginal bleeding for 4 months. DIAGNOSIS The patient was diagnosed with stage IC ovarian clear cell carcinoma that presented with a mutation of the phosphatidylinositol 4,5-bisphosphate 3-kinase alpha subunit (PIK3CA) gene. INTERVENTION We performed an early diagnosis and complete surgical resection of the tumor with platinum-based chemotherapy. OUTCOME This patient with mutation of the PIK3CA gene was sensitive to platinum-based chemotherapy, showed a significant downwards trend in tumor markers, and was in good health within the year of follow-up. LESSONS This study described an OCCC case that presented with a PIK3CA mutation and was successfully managed with careful and complete resection of the tumor. This patient with mutation of the PIK3CA gene was sensitive to platinum-based chemotherapy, showed a significant downwards trend in tumor markers, and did not have recurrence after a year of follow-up, indicating a reasonably good prognosis. Therefore, surgery plus platinum drug chemotherapy is still the best strategy for OCCC treatment. In addition, it is recommended for such patients to undergo genetic testing as much as possible to predict the clinical treatment effect.
Collapse
Affiliation(s)
- Abdulkarim Mohamed Farah
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | |
Collapse
|
47
|
Prevalence of ARID1A Mutations in Cell-Free Circulating Tumor DNA in a Cohort of 71,301 Patients and Association with Driver Co-Alterations. Cancers (Basel) 2022; 14:cancers14174281. [PMID: 36077815 PMCID: PMC9454642 DOI: 10.3390/cancers14174281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
ARID1A abnormalities disturb transcriptional processes regulated by chromatin remodeling and correlate with immunotherapy responsiveness. We report the first blood-based cell-free DNA (cfDNA) next-generation sequencing (NGS) ARID1A analysis. From November 2016 through August 2019, 71,301 patients with advanced solid tumors underwent clinical blood-derived cfDNA testing. Of these patients, 62,851 (88%) had ≥1 cfDNA alteration, and 3137 (of the 62,851) (5%) had ≥1 deleterious ARID1A alteration (a frequency similar to the ~6% generally reported in tissue NGS), suggesting this non-invasive test’s value in interrogating ARID1A. ARID1A cfDNA alterations were most frequent in endometrial cancer, 21.3% of patients; bladder cancer, 12.9%; gastric cancer, 11%; cholangiocarcinoma, 10.9%; and hepatocellular carcinoma, 10.6%. Blood samples with a functional ARID1A abnormality had more alterations/sample (median, 6 versus 4; p < 0.0001) and more frequent co-alterations in ≥1 gene in key oncogenic pathways: signal transduction, RAS/RAF/MAPK, PI3K/Akt/mTor, and the cell cycle. Taken together, our data suggest that liquid (blood) biopsies identify ARID1A alterations at a frequency similar to that found in primary tumor material. Furthermore, co-alterations in key pathways, some of which are pharmacologically tractable, occurred more frequently in samples with functional (deleterious) ARID1A alterations than in those without such aberrations, which may inform therapeutic strategies.
Collapse
|
48
|
Is There a Survival Benefit of Adjuvant Chemotherapy in Stage IC1 Epithelial Ovarian Cancer Patients? A Meta-Analysis. Curr Oncol 2022; 29:5763-5773. [PMID: 36005192 PMCID: PMC9406671 DOI: 10.3390/curroncol29080454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The purpose of the present systematic review is to clarify whether adjuvant chemotherapy improves survival rates in women with stage IC1 ovarian cancer. We searched Medline, Scopus, Clinicaltrials.gov, EMBASE, Cochrane Central Register of Controlled Trials CENTRAL and Google Scholar. We considered comparative observational studies and randomized trials that investigated survival outcomes (progression-free (PFS) and overall survival (OS)) among women with intraoperative rupture of early-stage epithelial ovarian cancer who received adjuvant chemotherapy and those that did not. Eleven studies, which recruited 7556 patients, were included. The risk of bias was defined as moderate after assessment with the Risk of Bias in non-Randomized Trials tool. Meta-analysis was performed with RStudio. Seven studies investigated the impact of adjuvant chemotherapy on recurrence-free survival of patients experiencing intraoperative cyst rupture for otherwise stage I ovarian cancer. The outcome was not affected by the use of adjuvant chemotherapy as the effect estimate was not significant (HR 1.24, 95% CI 0.74, 2.04). The analysis of data from 5 studies similarly revealed that overall survival rates were comparable among the two groups (HR 0.75, 95% CI 0.54, 1.05). This meta-analysis did not detect any benefit from adjuvant chemotherapy for stage IC ovarian cancer patients with cyst rupture. However, conclusions from this investigation are limited by a study population which included multiple histologic subtypes, high and low grade tumors and incompletely staged patients.
Collapse
|
49
|
Kim SI, Kim JH, Noh JJ, Kim SH, Kim TE, Kim K, Park JY, Lim MC, Lee JW, Kim JW. Impact of bevacizumab and secondary cytoreductive surgery on survival outcomes in platinum-sensitive relapsed ovarian clear cell carcinoma: A multicenter study in Korea. Gynecol Oncol 2022; 166:444-452. [PMID: 35863991 DOI: 10.1016/j.ygyno.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/30/2022] [Accepted: 07/10/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE This study investigated survival outcomes for platinum-sensitive relapsed ovarian clear cell carcinoma (OCCC) by treatment method. METHODS OCCC patients with platinum-sensitive recurrence that received secondary treatment at five institutions between July 2007 and June 2021 were included. Patient characteristics and survival outcomes were compared according to the use of bevacizumab (BEV) during second-line chemotherapy and secondary cytoreductive surgery (CRS). RESULTS 138 patients were included. The BEV (n = 36) and non-BEV (n = 102) groups had similar initial FIGO stages and proportions of secondary CRS. The BEV group showed improved progression-free survival (PFS; median, 15.4 vs. 7.5 months; P = 0.042) and overall survival (OS; P = 0.043) compared to the non-BEV group. In multivariate analyses, BEV was identified as an independent prognostic factor for PFS (adjusted hazard ratio [aHR], 0.571; 95% confidence interval [CI], 0.354-0.921; P = 0.022) and OS (aHR, 0.435; 95%CI, 0.195-0.970; P = 0.042). The secondary CRS group (n = 42) had early-stage disease at diagnosis more frequently (P = 0.009) and multi-site metastasis (P < 0.001) at recurrence less frequently than the no surgery group (n = 96). The secondary CRS group showed significantly better PFS (median, 33.7 vs. 7.2 months; P < 0.001) and OS (P < 0.001). Secondary CRS was associated with a significantly improved PFS (aHR, 0.297; 95% CI, 0.183-0.481; P < 0.001) and OS (aHR, 0.276; 95% CI, 0.133-0.576; P = 0.001). The BEV and non-BEV groups showed similar PFS and OS among the patients who underwent secondary CRS. In contrast, the BEV group showed improved PFS and OS among patients who did not undergo surgery. CONCLUSIONS The use of BEV during second-line chemotherapy and secondary CRS may improve PFS and OS in patients with platinum-sensitive relapsed OCCC. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Joseph J Noh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung-Ho Kim
- Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Eun Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jeong-Yeol Park
- Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
50
|
Ovarian Clear Cell Carcinoma with Functioning Stroma and Associated Endometrial Intraepithelial Neoplasia. Case Rep Obstet Gynecol 2022; 2022:5538390. [PMID: 35845973 PMCID: PMC9279104 DOI: 10.1155/2022/5538390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/04/2022] [Indexed: 11/18/2022] Open
Abstract
We report the case of a 79-year-old woman with a large pelvic mass and postmenopausal bleeding, associated with hyperestrogenism. A pelvic MRI shows the presence of a large mass of 12.6 cm originating from the right ovary without signs of metastasis. A total abdominal hysterectomy with unilateral salpingooophorectomy was performed, knowing the patient underwent a left salpingooophorectomy decades ago. The pathological findings showed an ovarian clear cell carcinoma (pT1A) with associated endometrial intraepithelial neoplasia. There is convincing evidence that the production of estrogen is located in the activated ovarian stroma. This supports the view that functioning stroma of ovarian cancer can lead to hyperestrogenism and eventually endometrial cancer.
Collapse
|